123 results on '"Sandya Liyanarachchi"'
Search Results
2. Supplementary Tables 1 - 4, Figures 1 - 6 from Dissection of the Major Hematopoietic Quantitative Trait Locus in Chromosome 6q23.3 Identifies miR-3662 as a Player in Hematopoiesis and Acute Myeloid Leukemia
- Author
-
Ann-Kathrin Eisfeld, Albert de la Chapelle, Clara D. Bloomfield, Denis C. Guttridge, James S. Blachly, Ramiro Garzon, Parvathi Ranganathan, Kevin W. Hoag, Malori A. Lankenau, Xiaomeng Huang, Maryam A. Bainazar, Mitra Patel, Sujay Mehta, Sandya Liyanarachchi, Christopher J. Walker, and Sophia E. Maharry
- Abstract
Supplementary Table 1. Linkage disequilibrium patterns of the tag-SNPs used for genotyping the 6q23.3 locus. Supplementary Table 2. Cytogenetic and molecular information on AML patients used for functional studies (n=12). Supplementary Table 3. French-American-British (FAB) classification35 and molecular information on AML patients used to determine miR-3662's abundance (n=8). Supplementary Table 4. Canonical pathway analysis of the miR-3662-associated gene expression signature. Supplementary Figure 1. Transcription factor binding according to the transcription factor chip data from ENCODE. Supplementary Figure 2. Electrophoretic mobility shift assay comparing the binding affinity of the alleles of rs66650371 and rs9483788. Supplementary Figure 3. Endogenous miR-3662 expression levels of hematopoietic progenitor (HP) cells during differentiation, total bone marrow aspirate of three non-leukemic donors (total BM 1-3), different populations of differentiated peripheral blood cells, and three AML cell lines. Supplementary Figure 4. Top panel, Macroscopic pictures of the spleens of three mice of the scramble and miR-3662-infected groups (organs harvested post-mortem). All mice had a massive splenomegaly compared to the un-injected, sacrificed control mouse. Bottom panel, images of spleen histologies (40x enlargement). Slides were stained for CD45 to proof MV4-11 origin of the leukemia. Supplementary Figure 5. Endogenous abundance of miR-3662 and IKBKB in patient samples and cell lines. Supplementary Figure 6. Comparison of the relative miR-3662 abundance of AML patient blasts and AML cell lines before (black) and after (red) forced miR-3662 expression with the lentiviral expression construct.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Data from Genetic Characterization and Prognostic Relevance of Acquired Uniparental Disomies in Cytogenetically Normal Acute Myeloid Leukemia
- Author
-
Clara D. Bloomfield, Albert de la Chapelle, John C. Byrd, Richard M. Stone, Andrew J. Carroll, Eunice S. Wang, Geoffrey L. Uy, Jonathan E. Kolitz, Bayard L. Powell, Kellie J. Archer, Shelley Orwick, Sophia E. Maharry, Luke K. Genutis, Brian Giacopelli, Christopher C. Oakes, Dimitrios Papaioannou, Marius Bill, James S. Blachly, Deedra Nicolet, Chi Song, Sandya Liyanarachchi, Krzysztof Mrózek, Ann-Kathrin Eisfeld, Jessica Kohlschmidt, and Christopher J. Walker
- Abstract
Supplementary Methods, Supplementary References Supplementary Table S1: Inherited regions of homozygosity in >1% of 1,798 non-leukemic individuals Supplementary Table S2: Acquired uniparental disomies detected in 425 cytogenetically normal AML patients Supplementary Table S3. Associations between recurrent UPDs with pretreatment patient characteristics for patients with cytogenetically normal acute myeloid leukemia Supplementary Table S5. Variant allele fraction of gene mutations that co-occurred with UPDs Supplementary Table S6. Allelic ratio of FLT3 internal tandem duplications that co-occurred with UPDs Supplementary Table S7: Copy number gains and losses in 425 patients with cytogenetically normal acute myeloid leukemia
- Published
- 2023
- Full Text
- View/download PDF
4. Data from Genetic Characterization and Prognostic Relevance of Acquired Uniparental Disomies in Cytogenetically Normal Acute Myeloid Leukemia
- Author
-
Clara D. Bloomfield, Albert de la Chapelle, John C. Byrd, Richard M. Stone, Andrew J. Carroll, Eunice S. Wang, Geoffrey L. Uy, Jonathan E. Kolitz, Bayard L. Powell, Kellie J. Archer, Shelley Orwick, Sophia E. Maharry, Luke K. Genutis, Brian Giacopelli, Christopher C. Oakes, Dimitrios Papaioannou, Marius Bill, James S. Blachly, Deedra Nicolet, Chi Song, Sandya Liyanarachchi, Krzysztof Mrózek, Ann-Kathrin Eisfeld, Jessica Kohlschmidt, and Christopher J. Walker
- Abstract
Purpose:Uniparental disomy (UPD) is a way cancer cells duplicate a mutated gene, causing loss of heterozygosity (LOH). Patients with cytogenetically normal acute myeloid leukemia (CN-AML) do not have microscopically detectable chromosome abnormalities, but can harbor UPDs. We examined the prognostic significance of UPDs and frequency of LOH in patients with CN-AML.Experimental Design: We examined the frequency and prognostic significance of UPDs in a set of 425 adult patients with de novo CN-AML who were previously sequenced for 81 genes typically mutated in cancer. Associations of UPDs with outcome were analyzed in the 315 patients with CN-AML younger than 60 years.Results:We detected 127 UPDs in 109 patients. Most UPDs were large and typically encompassed all or most of the affected chromosome arm. The most common UPDs occurred on chromosome arms 13q (7.5% of patients), 6p (2.8%), and 11p (2.8%). Many UPDs significantly cooccurred with mutations in genes they encompassed, including 13q UPD with FLT3-internal tandem duplication (FLT3-ITD; P < 0.001), and 11p UPD with WT1 mutations (P = 0.02). Among patients younger than 60 years, UPD of 11p was associated with longer overall survival (OS) and 13q UPD with shorter disease-free survival (DFS) and OS. In multivariable models that accounted for known prognostic markers, including FLT3-ITD and WT1 mutations, UPD of 13q maintained association with shorter DFS, and UPD of 11p maintained association with longer OS.Conclusions:LOH mediated by UPD is a recurrent feature of CN-AML. Detection of UPDs of 13q and 11p might be useful for genetic risk stratification of patients with CN-AML.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Table S4 from Genetic Characterization and Prognostic Relevance of Acquired Uniparental Disomies in Cytogenetically Normal Acute Myeloid Leukemia
- Author
-
Clara D. Bloomfield, Albert de la Chapelle, John C. Byrd, Richard M. Stone, Andrew J. Carroll, Eunice S. Wang, Geoffrey L. Uy, Jonathan E. Kolitz, Bayard L. Powell, Kellie J. Archer, Shelley Orwick, Sophia E. Maharry, Luke K. Genutis, Brian Giacopelli, Christopher C. Oakes, Dimitrios Papaioannou, Marius Bill, James S. Blachly, Deedra Nicolet, Chi Song, Sandya Liyanarachchi, Krzysztof Mrózek, Ann-Kathrin Eisfeld, Jessica Kohlschmidt, and Christopher J. Walker
- Abstract
Mutations detected in 425 CN-AML patients
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Prognostic DNA Methylation Biomarkers in Ovarian Cancer
- Author
-
Kenneth P. Nephew, Tim H-M. Huang, Sun Kim, Robert Brown, Gillian Gifford, Beth Y. Karlan, Ramana V. Davuluri, Joseph C. Wan, Zailong Wang, Lang Li, Sandya Liyanarachchi, Rae Lynn Baldwin, Yoo-Sung Kim, Henry H. Paik, Curtis Balch, and Susan H. Wei
- Abstract
Purpose: Aberrant DNA methylation, now recognized as a contributing factor to neoplasia, often shows definitive gene/sequence preferences unique to specific cancer types. Correspondingly, distinct combinations of methylated loci can function as biomarkers for numerous clinical correlates of ovarian and other cancers.Experimental Design: We used a microarray approach to identify methylated loci prognostic for reduced progression-free survival (PFS) in advanced ovarian cancer patients. Two data set classification algorithms, Significance Analysis of Microarray and Prediction Analysis of Microarray, successfully identified 220 candidate PFS-discriminatory methylated loci. Of those, 112 were found capable of predicting PFS with 95% accuracy, by Prediction Analysis of Microarray, using an independent set of 40 advanced ovarian tumors (from 20 short-PFS and 20 long-PFS patients, respectively). Additionally, we showed the use of these predictive loci using two bioinformatics machine-learning algorithms, Support Vector Machine and Multilayer Perceptron.Conclusion: In this report, we show that highly prognostic DNA methylation biomarkers can be successfully identified and characterized, using previously unused, rigorous classifying algorithms. Such ovarian cancer biomarkers represent a promising approach for the assessment and management of this devastating disease.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table 1 from Origins and Prevalence of the American Founder Mutation of MSH2
- Author
-
Albert de la Chapelle, Heather Hampel, Henry T. Lynch, Stephen N. Thibodeau, Brittany C. Thomas, Gordon Gong, Jane F. Lynch, Molly Deacon, Ilene Comeras, Victoria Schunemann, Dan Fix, Sandya Liyanarachchi, Kyle Walsh, Shuying Sun, Mark E. Baze, and Mark Clendenning
- Abstract
Supplementary Table 1 from Origins and Prevalence of the American Founder Mutation of MSH2
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Table 2 from A Susceptibility Locus for Papillary Thyroid Carcinoma on Chromosome 8q24
- Author
-
Albert de la Chapelle, Juha Kere, Saul Suster, Wei Li, Hong Jiao, Sandya Liyanarachchi, Rebecca Nagy, and Huiling He
- Abstract
Supplementary Table 2 from A Susceptibility Locus for Papillary Thyroid Carcinoma on Chromosome 8q24
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table 2 from Xenoestrogen-Induced Epigenetic Repression of microRNA-9-3 in Breast Epithelial Cells
- Author
-
Tim H-M. Huang, Alfred S.L. Cheng, Joseph Liu, Tao Zuo, Yu-I Weng, Sandya Liyanarachchi, Benjamin A.T. Rodriguez, Daniel E. Deatherage, and Pei-Yin Hsu
- Abstract
Supplementary Table 2 from Xenoestrogen-Induced Epigenetic Repression of microRNA-9-3 in Breast Epithelial Cells
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure 1 from A Susceptibility Locus for Papillary Thyroid Carcinoma on Chromosome 8q24
- Author
-
Albert de la Chapelle, Juha Kere, Saul Suster, Wei Li, Hong Jiao, Sandya Liyanarachchi, Rebecca Nagy, and Huiling He
- Abstract
Supplementary Figure 1 from A Susceptibility Locus for Papillary Thyroid Carcinoma on Chromosome 8q24
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figures 1-6, Tables 1-15, Methods from Epigenetic Silencing Mediated through Activated PI3K/AKT Signaling in Breast Cancer
- Author
-
Tim H.-M. Huang, Victor X. Jin, Alfred S. L. Cheng, Huey-Jen L. Lin, Charles L. Shapiro, Bhuvaneswari Ramaswamy, Cenny Taslim, Pei-Yin Hsu, Daniel E. Deatherage, Sandya Liyanarachchi, Yi-Wen Huang, Fei Gu, Rulong Shen, Yu-I Weng, Xun Lan, Ta-Ming Liu, and Tao Zuo
- Abstract
Supplementary Figures 1-6, Tables 1-15, Methods from Epigenetic Silencing Mediated through Activated PI3K/AKT Signaling in Breast Cancer
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 2 from A Susceptibility Locus for Papillary Thyroid Carcinoma on Chromosome 8q24
- Author
-
Albert de la Chapelle, Juha Kere, Saul Suster, Wei Li, Hong Jiao, Sandya Liyanarachchi, Rebecca Nagy, and Huiling He
- Abstract
Supplementary Figure 2 from A Susceptibility Locus for Papillary Thyroid Carcinoma on Chromosome 8q24
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Table 1 from A Susceptibility Locus for Papillary Thyroid Carcinoma on Chromosome 8q24
- Author
-
Albert de la Chapelle, Juha Kere, Saul Suster, Wei Li, Hong Jiao, Sandya Liyanarachchi, Rebecca Nagy, and Huiling He
- Abstract
Supplementary Table 1 from A Susceptibility Locus for Papillary Thyroid Carcinoma on Chromosome 8q24
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure 1 from Origins and Prevalence of the American Founder Mutation of MSH2
- Author
-
Albert de la Chapelle, Heather Hampel, Henry T. Lynch, Stephen N. Thibodeau, Brittany C. Thomas, Gordon Gong, Jane F. Lynch, Molly Deacon, Ilene Comeras, Victoria Schunemann, Dan Fix, Sandya Liyanarachchi, Kyle Walsh, Shuying Sun, Mark E. Baze, and Mark Clendenning
- Abstract
Supplementary Figure 1 from Origins and Prevalence of the American Founder Mutation of MSH2
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Table 1 from Xenoestrogen-Induced Epigenetic Repression of microRNA-9-3 in Breast Epithelial Cells
- Author
-
Tim H-M. Huang, Alfred S.L. Cheng, Joseph Liu, Tao Zuo, Yu-I Weng, Sandya Liyanarachchi, Benjamin A.T. Rodriguez, Daniel E. Deatherage, and Pei-Yin Hsu
- Abstract
Supplementary Table 1 from Xenoestrogen-Induced Epigenetic Repression of microRNA-9-3 in Breast Epithelial Cells
- Published
- 2023
- Full Text
- View/download PDF
16. Data from Breast Cancer–Associated Fibroblasts Confer AKT1-Mediated Epigenetic Silencing of Cystatin M in Epithelial Cells
- Author
-
Tim H.-M. Huang, Michael C. Ostrowski, Ann-Lii Cheng, Pearlly S. Yan, Shili Lin, Lisa Asamoto, Dustin Potter, Daniel E. Deatherage, Rulong Shen, Shuying Sun, Sandya Liyanarachchi, Chieh Ti Kuo, Ching-Hung Lin, Tao Zuo, and Huey-Jen L. Lin
- Abstract
The interplay between histone modifications and promoter hypermethylation provides a causative explanation for epigenetic gene silencing in cancer. Less is known about the upstream initiators that direct this process. Here, we report that the Cystatin M (CST6) tumor suppressor gene is concurrently down-regulated with other loci in breast epithelial cells cocultured with cancer-associated fibroblasts (CAF). Promoter hypermethylation of CST6 is associated with aberrant AKT1 activation in epithelial cells, as well as the disabled INNP4B regulator resulting from the suppression by CAFs. Repressive chromatin, marked by trimethyl-H3K27 and dimethyl-H3K9, and de novo DNA methylation is established at the promoter. The findings suggest that microenvironmental stimuli are triggers in this epigenetic cascade, leading to the long-term silencing of CST6 in breast tumors. Our present findings implicate a causal mechanism defining how tumor stromal fibroblasts support neoplastic progression by manipulating the epigenome of mammary epithelial cells. The result also highlights the importance of direct cell-cell contact between epithelial cells and the surrounding fibroblasts that confer this epigenetic perturbation. Because this two-way interaction is anticipated, the described coculture system can be used to determine the effect of epithelial factors on fibroblasts in future studies. [Cancer Res 2008;68(24):10257–66]
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Table 2 from Origins and Prevalence of the American Founder Mutation of MSH2
- Author
-
Albert de la Chapelle, Heather Hampel, Henry T. Lynch, Stephen N. Thibodeau, Brittany C. Thomas, Gordon Gong, Jane F. Lynch, Molly Deacon, Ilene Comeras, Victoria Schunemann, Dan Fix, Sandya Liyanarachchi, Kyle Walsh, Shuying Sun, Mark E. Baze, and Mark Clendenning
- Abstract
Supplementary Table 2 from Origins and Prevalence of the American Founder Mutation of MSH2
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Table 3 from A Susceptibility Locus for Papillary Thyroid Carcinoma on Chromosome 8q24
- Author
-
Albert de la Chapelle, Juha Kere, Saul Suster, Wei Li, Hong Jiao, Sandya Liyanarachchi, Rebecca Nagy, and Huiling He
- Abstract
Supplementary Table 3 from A Susceptibility Locus for Papillary Thyroid Carcinoma on Chromosome 8q24
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Methods, Figures 1-5, Tables 1-5 from Breast Cancer–Associated Fibroblasts Confer AKT1-Mediated Epigenetic Silencing of Cystatin M in Epithelial Cells
- Author
-
Tim H.-M. Huang, Michael C. Ostrowski, Ann-Lii Cheng, Pearlly S. Yan, Shili Lin, Lisa Asamoto, Dustin Potter, Daniel E. Deatherage, Rulong Shen, Shuying Sun, Sandya Liyanarachchi, Chieh Ti Kuo, Ching-Hung Lin, Tao Zuo, and Huey-Jen L. Lin
- Abstract
Supplementary Methods, Figures 1-5, Tables 1-5 from Breast Cancer–Associated Fibroblasts Confer AKT1-Mediated Epigenetic Silencing of Cystatin M in Epithelial Cells
- Published
- 2023
- Full Text
- View/download PDF
20. Data from Origins and Prevalence of the American Founder Mutation of MSH2
- Author
-
Albert de la Chapelle, Heather Hampel, Henry T. Lynch, Stephen N. Thibodeau, Brittany C. Thomas, Gordon Gong, Jane F. Lynch, Molly Deacon, Ilene Comeras, Victoria Schunemann, Dan Fix, Sandya Liyanarachchi, Kyle Walsh, Shuying Sun, Mark E. Baze, and Mark Clendenning
- Abstract
Large germline deletions within the mismatch repair gene MSH2 account for a significant proportion (up to 20%) of all deleterious mutations of this gene which are associated with Lynch syndrome. An exons 1 to 6 deletion of MSH2, originally reported in nine families, has been associated with a founding event within the United States, which genealogic studies had previously dated to 1727, and the number of present day carriers was estimated to be 18,981. Here, we report the development of a robust multiplex PCR which has assisted in the detection of 32 new families who carry the MSH2 American Founder Mutation (AFM). By offering testing to family members, 126 carriers of the AFM have been identified. Extensive genealogic studies have connected 27 of the 41 AFM families into seven extended pedigrees. These extended families have been traced back to around the 18th century without any evidence of further convergence between them. Characterization of the genomic sequence flanking the deletion and the identification of a common disease haplotype of between 0.6 and 2.3 Mb in all probands provides evidence for a common ancestor between these extended families. The DMLE+2.2 software predicts an age of ∼500 years (95% confidence interval, 425–625) for this mutation. Taken together, these data are suggestive of an earlier founding event than was first thought, which likely occurred in a European or a Native American population. The consequences of this finding would be that the AFM is significantly more frequent in the United States than was previously predicted. [Cancer Res 2008;68(7):2145–53]
- Published
- 2023
- Full Text
- View/download PDF
21. Transcriptome analysis discloses dysregulated genes in normal appearing tumor-adjacent thyroid tissues from patients with papillary thyroid carcinoma
- Author
-
W. G. Li, Matthew D. Ringel, Sandya Liyanarachchi, Pearlly S. Yan, Albert de la Chapelle, Daniel F. Comiskey, Huiling He, Ralf Bundschuh, Altan M. Turkoglu, and Pamela Brock
- Subjects
Male ,0301 basic medicine ,Cell biology ,endocrine system ,endocrine system diseases ,Carcinogenesis ,Science ,Thyroid Gland ,Apoptosis ,Pathogenesis ,Biology ,medicine.disease_cause ,Article ,Thyroid carcinoma ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,Genetics ,medicine ,Humans ,Thyroid cancer ,Aged ,Cell Proliferation ,Multidisciplinary ,Gene Expression Profiling ,Thyroid ,fungi ,Computational Biology ,Cancer ,Middle Aged ,Microarray Analysis ,medicine.disease ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Thyroid Cancer, Papillary ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,Female ,Lymphocytic Thyroiditis - Abstract
Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer. The molecular characteristics of histologically normal appearing tissue adjacent to the tumor (NAT) from PTC patients are not well characterized. The aim of this study was to characterize the global gene expression profile of NAT and compare it with those of normal and tumor thyroid tissues. We performed total RNA sequencing with fresh frozen thyroid tissues from a cohort of three categories of samples including NAT, normal thyroid (N), and PTC tumor (T). Transcriptome analysis shows that NAT presents a unique gene expression profile, which was not associated with sex or the presence of lymphocytic thyroiditis. Among the differentially expressed genes (DEGs) of NAT vs N, 256 coding genes and 5 noncoding genes have been reported as cancer genes involved in cell proliferation, apoptosis, and/or tumorigenesis. Bioinformatics analysis with Ingenuity Pathway Analysis software revealed that “Cancer, Organismal Injury and Abnormalities, Cellular Response to Therapeutics, and Cellular Movement” were major dysregulated pathways in the NAT tissues. This study provides improved insight into the complexity of gene expression changes in the thyroid glands of patients with PTC.
- Published
- 2021
22. OR09-2 The RCAN 1.4 Metastasis Progression Suppressor Gene is Hypermethylated at Intron 1 and Downregulated in Papillary Thyroid Cancer
- Author
-
Wei Li, Sandya Liyanarachchi, Neel Rajan, Matthew Ringel, and Tilak Khanal
- Subjects
Endocrinology, Diabetes and Metabolism - Abstract
Progressive metastasis is the proximate cause of cancer-related mortality for the majority of patients with solid tumors. Identifying both drivers, and gatekeepers of late stage progression (metastasis progression suppressors; MPS) is crucial to improve biomarkers and treatments. We previously identified Regulator of Calcineurin 1.4 (RCAN1.4) as a MPS gene in thyroid cancer that functions in part through overexpression of the cap-n-collar transcription factor NFE2L3 (NRF3). However, the mechanisms for RCAN1.4 loss in thyroid cancer, and whether or not this is reversible, had not be addressed. Because the RCAN 1.4 promoter contains CG rich regions reported to be hypermethyalated in other tissues, we hypothesized that RCAN 1.4 downregulation in thyroid cancer was similarly regulated. Studies were performed in both cell lines and de-identified human papillary thyroid cancer (PTC) tumor and normal tissue pairs from our Endocrine Neoplasia Repository tumor bank. Five thyroid cancer cell lines (TPC1, FTC133, BCPAP, C643, and 8505C) with different genetic drivers were selected based on low expression of RCAN 1.4. To determine if lines display methylation-regulated expression of RCAN 1.4, cells were treated with 1-10uM decitabine, a DNA methyl transferase inhibitor. In all cell lines, RCAN1.4 mRNA and protein levels increased with decitabine treatment, consistent with hypermethylation. Analysis of the literature and in silico analysis of the RCAN 1.4 gene identified CG rich regions as putative methylation targets in the proximal promoter and in intron 1. Using two different methods; 1) isolation of MseI-digestion DNA fragments using CpG MethylQuest beads followed by qPCR and 2) methylation-specific PCR (MSP) of bisulfite-modified DNA, hypermethylation of the intron 1 CG rich region was identified. There was no evidence of hypermethylation of the CG rich region of the proximal promoter. Methylation of the two regions also was assessed on 18 pairs PTC and adjacent normal tissues. Similar to the cell lines, hypermethylation was identified at intron 1 of RCAN 1.4 in PTC versus normal tissues. QRT-PCR results confirmed lower levels of RCAN1-4 mRNA in the PTC vs normal tissue. These samples also demonstrated increased NRF3 expression versus normal tissue. Taken together, these data demonstrate that RCAN 1.4 is downregulated in thyroid cancer cells and human PTCs by hypermethylation of CG rich regions in intron 1 and that this effect is reversible using demethylating agents. Presentation: Saturday, June 11, 2022 11:45 a.m. - 12:00 p.m.
- Published
- 2022
- Full Text
- View/download PDF
23. Variants in LRRC34 reveal distinct mechanisms for predisposition to papillary thyroid carcinoma
- Author
-
Isabella V. Hendrickson, Pamela Brock, Sandya Liyanarachchi, Albert de la Chapelle, Lianbo Yu, Huiling He, Mehek S. Sheikh, Luke K Genutis, and Daniel F. Comiskey
- Subjects
0301 basic medicine ,Linkage disequilibrium ,Gene knockdown ,Genome-wide association study ,Biology ,Penetrance ,Thyroid carcinoma ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Germline mutation ,030220 oncology & carcinogenesis ,Genetics ,Cancer research ,Missense mutation ,Gene ,Genetics (clinical) - Abstract
BackgroundPapillary thyroid carcinoma (PTC) demonstrates high heritability and a low somatic mutation burden relative to other cancers. Therefore, the genetic risk predisposing to PTC is likely due to a combination of low penetrance variants. A recent genome-wide association study revealed the association of PTC with a missense variant, rs6793295, at 3q26 in a gene called Leucine Repeat Rich Containing 34 (LRRC34).MethodsWe report the mechanisms of PTC risk at 3q26 using a combination of overexpression, mass spectroscopy, knockdown, transcriptome profiling, migration assays and genetic analysis.ResultsWe observed differential binding of wild-type and missense LRRC34 to RANBP1. Overexpression of missense LRRC34 reduced RanGTP levels and increased apoptosis. We also identified a second linkage disequilibrium (LD) block upstream of LRRC34 containing regulatory variants with allele-specific expression. Transcriptome profiling of LRRC34 knockdown cells showed changes in genes involved with cellular movement. LRRC34 knockdown reduced the migration of thyroid cancer cell lines. Lastly, we assessed the relative contribution of PTC risk from each locus using haplotype analysis.ConclusionsOur study demonstrates two separate mechanisms, one in G protein signalling and the other in transcriptional control, dictating PTC risk at 3q26 using both biochemical and genetic techniques.
- Published
- 2020
- Full Text
- View/download PDF
24. A Truncating Germline Mutation of TINF2 in Individuals with Thyroid Cancer or Melanoma Results in Longer Telomeres
- Author
-
Pamela Brock, Katherine E. Delap, Yanqiang Wang, W. G. Li, Huiling He, Sandya Liyanarachchi, Daniel F. Comiskey, Albert de la Chapelle, and Taina T. Nieminen
- Subjects
Genetics ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,TINF2 ,Biology ,Shelterin ,TERF1 ,Frameshift mutation ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Germline mutation ,030220 oncology & carcinogenesis ,Mutation (genetic algorithm) ,Missense mutation ,Exome sequencing - Abstract
Background: Our genome sequencing analysis revealed a frameshift mutation in the shelterin gene TINF2 in a large family with individuals affected with papillary thyroid carcinoma (PTC) and melanoma. Here, we further characterized the mutation and screened for coding variants in the 6 shelterin genes in 24 families. Methods: Sanger sequencing was performed to screen for the TINF2 mutation in the key family. Quantitative reverse transcription-polymerase chain reaction (PCR) was used for TINF2 gene expression analysis. Exogenous expression and co-immunoprecipitation techniques were used for assessing TINF2 binding to TERF1. Relative telomere length (RTL) was quantified in DNAs from lymphocytes by using quantitative real-time PCR. Whole exome sequencing (WES) was performed in seven families with individuals affected with PTC and other cancer types. Screening for DNA variants in shelterin genes was performed by using whole genome sequencing data from 17 families and WES data from 7 further families. Results: The TINF2 mutation (TINF2 p.Trp198fs) showed complete co-segregation with PTC and melanoma in the key family. The mutation is not reported in databases and not identified in 23 other families we screened. The expression of TINF2 was borderline reduced in individuals with the mutation. The truncated TINF2 protein showed abolished binding to TERF1. The RTL in the individuals with the mutation was significantly longer when compared with those without the mutation from the same family as well as compared with 62 healthy controls. Among the 24 families, we identified 3 missense and 1 synonymous variant(s) in 2 shelterin genes (TINF2 and ACD). Conclusions: The rare frameshift mutation in the TINF2 gene and the associated longer telomere length suggest that dysregulated telomeres could be a mechanism predisposing to PTC and melanoma. DNA coding variants in shelterin genes are rare. Further studies are required to evaluate the roles of variants in shelterin genes in thyroid cancer and melanoma.
- Published
- 2020
- Full Text
- View/download PDF
25. Author Correction: GWAS of thyroid stimulating hormone highlights the pleiotropic effects and inverse association with thyroid cancer
- Author
-
Wei Zhou, Ben Brumpton, Omer Kabil, Julius Gudmundsson, Gudmar Thorleifsson, Josh Weinstock, Matthew Zawistowski, Jonas B. Nielsen, Layal Chaker, Marco Medici, Alexander Teumer, Silvia Naitza, Serena Sanna, Ulla T. Schultheiss, Anne Cappola, Juha Karjalainen, Mitja Kurki, Morgan Oneka, Peter Taylor, Lars G. Fritsche, Sarah E. Graham, Brooke N. Wolford, William Overton, Humaira Rasheed, Eirin B. Haug, Maiken E. Gabrielsen, Anne Heidi Skogholt, Ida Surakka, George Davey Smith, Anita Pandit, Tanmoy Roychowdhury, Whitney E. Hornsby, Jon G. Jonasson, Leigha Senter, Sandya Liyanarachchi, Matthew D. Ringel, Li Xu, Lambertus A. Kiemeney, Huiling He, Romana T. Netea-Maier, Jose I. Mayordomo, Theo S. Plantinga, Jon Hrafnkelsson, Hannes Hjartarson, Erich M. Sturgis, Aarno Palotie, Mark Daly, Cintia E. Citterio, Peter Arvan, Chad M. Brummett, Michael Boehnke, Albert de la Chapelle, Kari Stefansson, Kristian Hveem, Cristen J. Willer, and Bjørn Olav Åsvold
- Subjects
Multidisciplinary ,Science ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
The original version of this article contained an error in the results, in the second paragraph of the subsection entitled “Fine-mapping for potentially causal variants among TSH loci”, in which effect sizes for two variants were incorrectly reported.
- Published
- 2021
- Full Text
- View/download PDF
26. Genetic Characterization and Prognostic Relevance of Acquired Uniparental Disomies in Cytogenetically Normal Acute Myeloid Leukemia
- Author
-
Sophia E. Maharry, Clara D. Bloomfield, Marius Bill, Sandya Liyanarachchi, Christopher C. Oakes, Chi Song, Christopher J. Walker, Albert de la Chapelle, Geoffrey L. Uy, Brian Giacopelli, James S. Blachly, Bayard L. Powell, Eunice S. Wang, Shelley Orwick, Jonathan E. Kolitz, Richard Stone, Deedra Nicolet, Kellie J. Archer, Ann-Kathrin Eisfeld, John C. Byrd, Luke K Genutis, Jessica Kohlschmidt, Dimitrios Papaioannou, Krzysztof Mrózek, and Andrew J. Carroll
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Adolescent ,Loss of Heterozygosity ,medicine.disease_cause ,Disease-Free Survival ,Article ,Loss of heterozygosity ,Cytogenetics ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Aged ,Mutation ,business.industry ,Nuclear Proteins ,Chromosome ,Cancer ,Middle Aged ,Uniparental Disomy ,Prognosis ,medicine.disease ,Uniparental disomy ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Tandem exon duplication ,business - Abstract
Purpose:Uniparental disomy (UPD) is a way cancer cells duplicate a mutated gene, causing loss of heterozygosity (LOH). Patients with cytogenetically normal acute myeloid leukemia (CN-AML) do not have microscopically detectable chromosome abnormalities, but can harbor UPDs. We examined the prognostic significance of UPDs and frequency of LOH in patients with CN-AML.Experimental Design: We examined the frequency and prognostic significance of UPDs in a set of 425 adult patients with de novo CN-AML who were previously sequenced for 81 genes typically mutated in cancer. Associations of UPDs with outcome were analyzed in the 315 patients with CN-AML younger than 60 years.Results:We detected 127 UPDs in 109 patients. Most UPDs were large and typically encompassed all or most of the affected chromosome arm. The most common UPDs occurred on chromosome arms 13q (7.5% of patients), 6p (2.8%), and 11p (2.8%). Many UPDs significantly cooccurred with mutations in genes they encompassed, including 13q UPD with FLT3-internal tandem duplication (FLT3-ITD; P < 0.001), and 11p UPD with WT1 mutations (P = 0.02). Among patients younger than 60 years, UPD of 11p was associated with longer overall survival (OS) and 13q UPD with shorter disease-free survival (DFS) and OS. In multivariable models that accounted for known prognostic markers, including FLT3-ITD and WT1 mutations, UPD of 13q maintained association with shorter DFS, and UPD of 11p maintained association with longer OS.Conclusions:LOH mediated by UPD is a recurrent feature of CN-AML. Detection of UPDs of 13q and 11p might be useful for genetic risk stratification of patients with CN-AML.
- Published
- 2019
- Full Text
- View/download PDF
27. Risk Haplotypes Uniquely Associated with Radioiodine-Refractory Thyroid Cancer Patients of High African Ancestry
- Author
-
Brynn Hollingsworth, Jennifer A. Sipos, Luke K Genutis, Yi Seok Chang, Christopher J. Walker, Pamela Brock, Electron Kebebew, Patience Green, Fadi Nabhan, W. G. Li, Sandya Liyanarachchi, Gilbert J. Cote, Steven I. Sherman, Sissy M. Jhiang, Albert de la Chapelle, Shuai Xue, Zachary A. Hurst, Huiling He, and Eric Menq
- Subjects
Adult ,Male ,Proto-Oncogene Proteins B-raf ,Oncology ,medicine.medical_specialty ,Adolescent ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Disease ,Polymorphism, Single Nucleotide ,Radiation Tolerance ,Risk Assessment ,Thyroglobulin ,White People ,Iodine Radioisotopes ,Ligases ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Refractory ,Risk Factors ,hemic and lymphatic diseases ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Genetic Predisposition to Disease ,Thyroid Neoplasms ,Thyroid cancer ,Germ-Line Mutation ,Aged ,African american ,BRCA1 Protein ,business.industry ,Incidence ,Haplotype ,Thyroid Cancer and Nodules ,Middle Aged ,medicine.disease ,United States ,Black or African American ,Phenotype ,Haplotypes ,030220 oncology & carcinogenesis ,Female ,Radiopharmaceuticals ,business - Abstract
Background: Thyroid cancer patients with radioiodine-refractory (RAI-R) disease, resulting from insufficient RAI delivery and/or RAI resistance, have increased mortality and limited treatment options. To date, studies have largely focused on tumor mutations associated with different stages of disease, which could provide prognostic value for RAI-R disease. It was hypothesized that germline variants contributing to intrinsic differences in iodine metabolism, tumor microenvironment, and/or immune surveillance are associated with RAI-R disease. Methods: Whole-genome genotyping data analysis was performed on 1145 Caucasian (CAU) patients, 244 of whom were RAI-R, and 55 African American (AA) patients, nine of whom were RAI-R. Germline-variant association studies were conducted using candidate genes involved in iodine metabolism or DNA-damage repair, as well as genome-wide association analysis. Initial data indicated several notable variants in a small number of patients (n = 7), who were later determined to be AA patients of >80% African ancestry (n = 37). This led to the study focusing on germline single nucleotide polymorphisms uniquely associated with RAI-R AA patients. Sanger sequencing was performed to validate risk alleles and identify the incidence of the common somatic mutations BRAF(V600E), NRAS(Q61R), and HRAS(Q61R) in AA patients whose primary tumor samples were available (28/55). Results: TG, BRCA1, and NSMCE2 haplotypes were identified as being uniquely associated with RAI-R AA patients of >80% African ancestry. All patients with the TG haplotype (n = 4) had a biochemical incomplete response to RAI therapy. Patients with the NSMCE2 haplotype (n = 4) were diagnosed at a young age (13, 17, 17, and 26 years old) with distant metastatic disease at initial diagnosis. The BRCA1 haplotype co-occurred in three out of four patients with the NSMCE2 haplotype. The incidence of BRAF(V600E) appears lower in papillary thyroid carcinomas from AA patients of >80% African ancestry (3/14; 21%) than in AA patients of
- Published
- 2019
- Full Text
- View/download PDF
28. Clinical characteristics of patients with colorectal cancer with double somatic mismatch repair mutations compared with Lynch syndrome
- Author
-
Rachel Pearlman, Sandya Liyanarachchi, Heather Hampel, Albert de la Chapelle, Wendy L. Frankel, Sigurdis Haraldsdottir, Colin C. Pritchard, Kari Stefansson, Thorunn Rafnar, and Jon G. Jonasson
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Colorectal cancer ,Population ,Microsatellite instability ,MLH1 ,medicine.disease ,Lynch syndrome ,MSH6 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Germline mutation ,030220 oncology & carcinogenesis ,Internal medicine ,Genetics ,medicine ,PMS2 ,business ,education ,Genetics (clinical) - Abstract
BackgroundPatients with colorectal cancer (CRC) with mismatch repair-deficient (dMMR) tumours without MLH1 methylation or germline MMR pathogenic variants (PV) were previously thought to have Lynch syndrome (LS). It is now appreciated that they can have double somatic (DS) MMR PVs. We explored the clinical characteristics between patients with DS tumours and LS in two population-based cohorts.MethodsWe included patients with CRC from Ohio 2013–2016 and Iceland 2000–2009. All had microsatellite instability testing and/or immunohistochemistry (IHC) of MMR proteins, and MLH1 methylation testing when indicated. Germline next-generation sequencing was performed for all with dMMR tumours; tumour sequencing followed for patients with unexplained dMMR. Clinical characteristics of DS patients and patients with LS were compared.ResultsOf the 232 and 51 patients with non-methylated dMMR tumours in the Ohio and Iceland cohorts, respectively, 57.8% (n=134) and 45.1% (n=23) had LS, 32.8% (n=76) and 31.4% (n=16) had DS PVs, 6% (n=14) and 9.8% (n=5) were unexplained and 4.3% (n=10) and 13.7% (n=7) had incorrect IHC. Age of diagnosis for DS patients was older than patients with LS (p=3.73×10−4) in the two cohorts. Patients with LS were more likely to meet Amsterdam II criteria (OR=15.81, p=8.47×10−6) and have multiple LS-associated tumours (OR=6.67, p=3.31×10−5). Absence of MLH1/PMS2 was predictive of DS PVs; isolated MSH6 and PMS2 absence was predictive of LS in both cohorts.ConclusionsIndividuals with LS are 15× more likely to meet Amsterdam II criteria and >5× more likely to have multiple cancers as compared with those with DS tumours. Furthermore, isolated loss of MSH6 or PMS2 protein predicts LS.
- Published
- 2019
- Full Text
- View/download PDF
29. A novel essential splice site variant in SPTB in a large hereditary spherocytosis family
- Author
-
Pamela Brock, Taina T. Nieminen, Huiling He, Yanqiang Wang, Daniel F. Comiskey, Isabella V. Hendrickson, W. G. Li, Sandya Liyanarachchi, Albert de la Chapelle, Department of Medical and Clinical Genetics, and Biosciences
- Subjects
0301 basic medicine ,Adult ,Male ,Heterozygote ,Adolescent ,Spherocytosis ,education ,Locus (genetics) ,Spherocytosis, Hereditary ,DNA variant ,030105 genetics & heredity ,Biology ,QH426-470 ,Clinical Reports ,Frameshift mutation ,Hereditary spherocytosis ,03 medical and health sciences ,symbols.namesake ,Exon ,splicing ,medicine ,Genetics ,Humans ,Spectrin ,hereditary spherocytosis ,Child ,Molecular Biology ,Genetics (clinical) ,Sanger sequencing ,Clinical Report ,1184 Genetics, developmental biology, physiology ,Spectrin repeat ,medicine.disease ,SPTB ,Pedigree ,030104 developmental biology ,Phenotype ,Mutation ,symbols ,Female ,RNA Splice Sites - Abstract
Background We studied a large family with 22 individuals affected with autosomal dominant hereditary spherocytosis (HS). Methods Genome‐wide linkage, whole‐genome sequencing (WGS), Sanger sequencing, RT‐PCR, and ToPO TA cloning analyses were performed. Results We revealed a heterozygous G>A transition in the 14q23 locus, at position +1 of the intron 8 donor splice site of the spectrin beta, erythrocytic (SPTB) gene. This splice variant (SPTB c.1064+1G>A) was confirmed by Sanger sequencing and showed complete co‐segregation with HS in the family. Further RT‐PCR reactions and sequencing analysis indicated that the variant leads to the exclusion of exon 8 and subsequent frameshift in exon 9 and a premature stop codon in SPTB. Translation of the altered allele would lead to a truncation with a loss of all spectrin repeat domains in SPTB protein. Conclusion This variant is novel and has not been found in any databases. We propose that this splice variant explains the spherocytosis phenotype observed in this large family., We studied a large family with 22 individuals affected with autosomal dominant hereditary spherocytosis (HS). Genome‐wide linkage and whole‐genome sequencing analyses revealed a heterozygous G>A transition in the 14q23 locus, at position +1 of the intron 8 donor splice site of the spectrin beta, erythrocytic (SPTB) gene.
- Published
- 2021
30. Multiethnic genome-wide association study of differentiated thyroid cancer in the EPITHYR consortium
- Author
-
Pierre Laurent-Puig, Yan Ren, Frédérique Rachédi, Pascal Guénel, Sandya Liyanarachchi, Asta Försti, Albert de la Chapelle, Om Kulkarni, Claire Mulot, Huiling He, Federica Gemignani, Julie Guibon, Anne-Valérie Guizard, Fabienne Lesueur, Rossella Elisei, Ausrele Kesminiene, Anthony F. Herzig, Mojgan Karimi, Pierre-Emmanuel Sugier, Thérèse Truong, Delphine Bacq-Daian, Anne-Louise Leutenegger, Constance Xhaard, Daniel F. Comiskey, Celia M Pereda, Evgenia Ostroumova, Florent de Vathaire, Elisabeth Adjadj, Françoise Borson-Chazot, Jean-François Deleuze, Carole Rubino, Anne Boland-Auge, Hauke Thomsen, Elise A. Lucotte, Marie-Christine Boutron-Ruault, Juan J Lence-Anta, Rosa Ortiz, Claire Schvartz, Institut Gustave Roussy (IGR), Centre de recherche en épidémiologie et santé des populations (CESP), Université de Versailles Saint-Quentin-en-Yvelines (UVSQ)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Paul Brousse-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay, Université Paris-Saclay, Cancer et génome: Bioinformatique, biostatistiques et épidémiologie d'un système complexe, Institut Curie [Paris]-MINES ParisTech - École nationale supérieure des mines de Paris, Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM), Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Épidémiologie des radiations, épidémiologie clinique des cancers et survie (U1018 (Équipe 3) ), Institut Gustave Roussy (IGR)-Centre de recherche en épidémiologie et santé des populations (CESP), Université de Versailles Saint-Quentin-en-Yvelines (UVSQ)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Paul Brousse-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay-Université de Versailles Saint-Quentin-en-Yvelines (UVSQ)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Paul Brousse-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay, Centre d'investigation clinique plurithématique Pierre Drouin [Nancy] (CIC-P), Centre d'investigation clinique [Nancy] (CIC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL)-Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Cardiovascular and Renal Clinical Trialists [Vandoeuvre-les-Nancy] (INI-CRCT), Institut Lorrain du Coeur et des Vaisseaux Louis Mathieu [Nancy], French-Clinical Research Infrastructure Network - F-CRIN [Paris] (Cardiovascular & Renal Clinical Trialists - CRCT ), Service de biostatistique et d'épidémiologie (SBE), Direction de la recherche clinique [Gustave Roussy], Institut Gustave Roussy (IGR)-Institut Gustave Roussy (IGR), Centre National de Recherche en Génomique Humaine (CNRGH), Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Université de Paris (UP), Université Paris Diderot, Sorbonne Paris Cité, Paris, France, Université Paris Diderot - Paris 7 (UPD7), Institut Jean Godinot [Reims], UNICANCER, Registre Général des Tumeurs du Calvados, CHU Caen, Normandie Université (NU)-Tumorothèque de Caen Basse-Normandie (TCBN)-Normandie Université (NU)-Tumorothèque de Caen Basse-Normandie (TCBN)-Centre Régional de Lutte contre le Cancer François Baclesse [Caen] (UNICANCER/CRLC), Normandie Université (NU)-UNICANCER-Tumorothèque de Caen Basse-Normandie (TCBN)-UNICANCER, Unité de recherche interdisciplinaire pour la prévention et le traitement des cancers (ANTICIPE), Université de Caen Normandie (UNICAEN), Normandie Université (NU)-Normandie Université (NU)-CHU Caen, Normandie Université (NU)-Tumorothèque de Caen Basse-Normandie (TCBN)-Tumorothèque de Caen Basse-Normandie (TCBN)-Centre Régional de Lutte contre le Cancer François Baclesse [Caen] (UNICANCER/CRLC), Normandie Université (NU)-UNICANCER-Tumorothèque de Caen Basse-Normandie (TCBN)-UNICANCER-Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital de Taaone, Hôpital Louis Pradel [CHU - HCL], Hospices Civils de Lyon (HCL), Health Service and Performance Research (HESPER), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon, Institute of Oncology and Radiobiology, Ohio State University [Columbus] (OSU), University of Pisa - Università di Pisa, Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ), Hopp Children's Cancer Center Heidelberg [Heidelber, Germany] (KITZ), German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ)-Heidelberg University Hospital [Heidelberg], German Cancer Consortium [Heidelberg] (DKTK), Génétique, génomique fonctionnelle et biotechnologies (UMR 1078) (GGB), Institut Brestois Santé Agro Matière (IBSAM), Université de Brest (UBO)-Université de Brest (UBO)-EFS-Institut National de la Santé et de la Recherche Médicale (INSERM), Maladies neurodéveloppementales et neurovasculaires (NeuroDiderot (UMR_S_1141 / U1141)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), Centre International de Recherche contre le Cancer - International Agency for Research on Cancer (CIRC - IARC), Organisation Mondiale de la Santé / World Health Organization Office (OMS / WHO), Ministère de l'Enseignement supérieur, de la Recherche et de l'Innovation, MESRI: 2102 918823 National Institute of Ecology, NIE Agence Nationale de la Recherche, ANR: ANR‐10‐COHO‐0006 Fondation ARC pour la Recherche sur le Cancer, ARC: PGA120150202302 Ligue Contre le Cancer Fondation de France Institut National Du Cancer, INCa: 9533 Agence Nationale de Sécurité Sanitaire de l’Alimentation, de l’Environnement et du Travail, ANSES Mutuelle Générale de l'Education Nationale, MGEN Electricité de France, EDF: EP 2019‐01, We thank Dr Yannick Rougier and Dr Dominique Baron-Dubourdieu for providing pathological reports for the NC study, as well as Dr Sylvie Laumond, Dr Jean-Paul Grangeon (Direction des affaires sanitaires et sociales de Nouvelle-Cal?donie) and the country's provincial health authorities (DPASS Sud, DPASS Nord, DPASS Iles Loyaut?) for support during data collection in the NC study. We thank Milagros Velasco, Mae Chappe and Idalmis Infante (Institute of Oncology and Radiobiology, La Havana, Cuba) and Silvia Turcios (National Institute of Endocrinology, La Havana, Cuba) for helping in the collection of the data in the Cuban Study. We thank John Paoaafaite and Joseph Teuri who contacted and interviewed cases and controls for the study. Finally, we also thank P. Morales, J. Iltis, P. Giraud, P. Didiergeorge, M. Brisard, G. Soubiran, B. Caillou, P. Dupire, J. Ienfa, G. de Clermont, N. Cerf, B. Oddo, M. Bambridge, C. Baron, A. Mouchard-Rachet, O. Simonet, D. Lamarque, J. Vabret, J. Delacre, M.P. Darquier and J. Leninger, for their help in the collection of the cases or in the organization of study in French Polynesia. We would like to thank the Association Centre de Regroupement Informatique et Statistique en Anatomie Pathologique en Provence-Alpes-C?te d'Azur (CRISAP PACA), as well as Dr Arlette Danzon, Dr Genevi?ve Sasolas, Dr Marc Christophe Sattonnet, Dr Marc Colonna, Dr Brigitte Lacour, Dr Michel Velten, Dr Enora Clero, Dr St?phane Maillard, Dr Laurent Bailly, Dr Eug?nia Marin? Barjoan, Dr Jean-Luc Lassalle, Dr Z Hafdi-Nejjari, Dr P Delafosse, Dr Elisabeth Adjadj, Kami-Marie Moreau, Cyrielle Orenes, Laurianne Sarrazin, St?phanie Bonnay, Fr?d?rique Chatelain, Maryse Barouh, Evelyne Rapp, Julie Festra?ts, Julie Valbousquet, Yusuf Atilgan, Jean Chappellet, Lallia Bedhouche, Florent Dayet and Ziyan Fami, for their help in the collection of cases, the organization and the management of the Young-Thyr study. We acknowledge Stefano Landi for the Italian GWAS data and Subhayan Chattopadhyay and Yasmeen Niazi (Division of Molecular Genetic Epidemiology, German Cancer Research Center?DKFZ) for technical assistance in these data analysis. We are grateful to Dr Herv? Perdry for his help in using the GASTON package. The EPITHYR GWAS was supported by Institut National du Cancer (grant number 9533) and Fondation ARC (grant number PGA120150202302). The E3N cohort received support from the MGEN, Gustave Roussy and Ligue contre le cancer for its setup and maintenance. The E3N cohort was also supported by a state grant from the Agence Nationale pour la Recherche (ANR) (grant number ANR-10-COHO-0006) within the Investissement d'Avenir program and from the French Ministry of Higher Education, Research and Innovation (MESRI, grant number 2102 918823). The other participating studies were funded by Ligue Nationale Contre le Cancer, ANR, the Direction G?n?rale de la Sante, the Agence Fran?aise de S?curit? Sanitaire de l'alimentation, de l'environnement et du travail (ANSES), CHILDTHYR EEC program, and the Fondation de France. JG was the recipient of a PhD fellowship from R?gion Ile-de-France, part of OK was the recipient of a post-doc fellowship from Electricit? de France (conseil scientifique de Radioprotection d'EDF, grant EP 2019-01)., The EPITHYR GWAS was supported by Institut National du Cancer (grant number 9533) and Fondation ARC (grant number PGA120150202302). The E3N cohort received support from the MGEN, Gustave Roussy and Ligue contre le cancer for its setup and maintenance. The E3N cohort was also supported by a state grant from the Agence Nationale pour la Recherche (ANR) (grant number ANR‐10‐COHO‐0006) within the Investissement d'Avenir program and from the French Ministry of Higher Education, Research and Innovation (MESRI, grant number 2102 918823). The other participating studies were funded by Ligue Nationale Contre le Cancer, the Direction Générale de la Sante, the Agence Française de Sécurité Sanitaire de l'alimentation, and the Fondation de France. JG was the recipient of a PhD fellowship from Région Ile‐de‐France, part of OK was the recipient of a post‐doc fellowship from Electricité de France (conseil scientifique de Radioprotection d'EDF, grant EP 2019‐01)., Mines Paris - PSL (École nationale supérieure des mines de Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Université Paris Cité (UPCité), EFS-Université de Brest (UBO)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Brestois Santé Agro Matière (IBSAM), Université de Brest (UBO), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité)
- Subjects
Male ,Cancer Research ,Native Hawaiian or Other Pacific Islander ,[SDV]Life Sciences [q-bio] ,Ethnic group ,Genome-wide association study ,0302 clinical medicine ,Gene Frequency ,Informed consent ,Epidemiology ,thyroid cancer ,Chromosomes, Human ,Medicine ,Prospective cohort study ,Thyroid cancer ,Genetics ,0303 health sciences ,education.field_of_study ,Incidence (epidemiology) ,Middle Aged ,Checklist ,3. Good health ,Oncology ,030220 oncology & carcinogenesis ,Pacific islanders ,Female ,Adult ,medicine.medical_specialty ,case-control study ,education ,Population ,Biology ,Pacific Islands ,Polymorphism, Single Nucleotide ,White People ,03 medical and health sciences ,Humans ,Genetic Predisposition to Disease ,Thyroid Neoplasms ,Aged ,030304 developmental biology ,genome-wide association study ,business.industry ,Case-control study ,medicine.disease ,Clinical trial ,Case-Control Studies ,Family medicine ,business - Abstract
Incidence of differentiated thyroid carcinoma (DTC) varies considerably between ethnic groups, with particularly high incidence rates in Pacific Islanders. Here, we conducted a genome-wide association study (GWAS) involving 1,554 cases/1,973 controls of European ancestry and 301 cases/348 controls of Oceanian ancestry from the EPITHYR consortium. Our results confirmed the association with the known DTC susceptibility loci at 2q35, 8p12, 9q22.33 and 14q13.3 in the European ancestry population and suggested two novel signals at 1p31.3 (rs334729) and 16q23.2 (rs16950982), which were associated with TSH levels in previous GWAS. We additionally replicated an association with 5p15.33 reported previously in Chinese and European populations. Except at 1p31.3, all associations were in the same direction in the population of Oceanian ancestry. The frequency of risk alleles at 2q35, 5p15.33 and 16q23.2 were significantly higher in Oceanians than in Europeans and may explain part of the highest DTC incidence observed in Oceanians.Competing Interest StatementThe authors have declared no competing interest.Funding StatementINCA (grant number 9533) and ARC (grant number PGA120150202302), Ligue Nationale Contre le Cancer, Agence Nationale pour la Recherche (ANR), the Direction Generale de la Sante, the Agence Francaise de Securite Sanitaire de l alimentation, de lenvironnement et du travail (ANSES), CHILDTHYR EEC program, and the Fondation de France. JG and CX were the recipient of a PhD fellowship from Region Ile-de-France.Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:All participants provided informed consent and each study was approved by their governing ethics committee.All necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).Yes I have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesThe data that support the findings of this study are available on request from the corresponding author. The data are not publicly available due to privacy or ethical restrictions. (Less)
- Published
- 2021
- Full Text
- View/download PDF
31. Characterizing the function of EPB41L4A in the predisposition to papillary thyroid carcinoma
- Author
-
Mehek S. Sheikh, Huiling He, Daniel F. Comiskey, Isabella V. Hendrickson, Sandya Liyanarachchi, Lianbo Yu, Pamela Brock, and Albert de la Chapelle
- Subjects
endocrine system diseases ,Genotype ,lcsh:Medicine ,Locus (genetics) ,Biology ,Mitochondrial Membrane Transport Proteins ,Article ,Thyroid cancer ,Thyroid carcinoma ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Thyroid Neoplasms ,lcsh:Science ,Enhancer ,Wnt Signaling Pathway ,Alleles ,Gene knockdown ,Multidisciplinary ,Microarray analysis techniques ,lcsh:R ,Alternative splicing ,medicine.disease ,Carcinoma, Papillary ,Haplotypes ,Thyroid Cancer, Papillary ,RNA splicing ,Cancer research ,lcsh:Q - Abstract
Papillary thyroid carcinoma (PTC) is the most common histotype of thyroid carcinoma. The heritability of PTC is high compared to other cancers, but its underlying causes are unknown. A recent genome-wide association study revealed the association of a variant at the 5q22 locus, rs73227498, with PTC predisposition. We report that rs17134155, a variant in high linkage disequilibrium with rs73227498, is located in an enhancer region downstream of coding transcripts of EPB41L4A. Rs17134155 showed significant enhancer activity in luciferase assays, and haplotypes containing the protective allele of this variant conferred a significantly lower risk of PTC. While the index SNP, rs73227498, acted as a significant cis-eQTL for expression of EPB41L4A, rs17134155 was a significant cis-sQTL for the alternative splicing of a non-coding transcript of EPB41L4A, called EPB41L4A-203. We also performed knockdown of EPB41L4A followed by microarray analysis. Some of the top differentially-expressed genes were represented among regulators of the WNT/β-catenin signaling pathway. Our results indicate that an enhancer region at 5q22 regulates the expression and splicing of EPB41L4A transcripts. We also provide evidence that EPB41L4A expression is involved in regulating growth and differentiation pathways, suggesting that decreased expression of EPB41L4A is a mechanism in the predisposition to PTC.
- Published
- 2020
- Full Text
- View/download PDF
32. Fine mapping of 14q13 reveals novel variants associated with different histological subtypes of papillary thyroid carcinoma
- Author
-
Krzysztof Sworczak, Leigha Senter, Andrew Thomas, Jarosław Jendrzejewski, Sandya Liyanarachchi, Albert de la Chapelle, Andrew Eiterman, Huiling He, and Rebecca Nagy
- Subjects
Male ,Cancer Research ,endocrine system diseases ,Single-nucleotide polymorphism ,Locus (genetics) ,Genome-wide association study ,Biology ,Polymorphism, Single Nucleotide ,Article ,Thyroid carcinoma ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Genotype ,Genetic predisposition ,Humans ,Genetic Predisposition to Disease ,Thyroid Neoplasms ,Allele frequency ,Chromosomes, Human, Pair 14 ,Gene Expression Profiling ,Chromosome Mapping ,Molecular biology ,Phenotype ,Oncology ,Thyroid Cancer, Papillary ,030220 oncology & carcinogenesis ,Female ,Genome-Wide Association Study - Abstract
The first two genome wide association studies (GWAS) of papillary thyroid carcinoma (PTC) detected five variants associated with PTC. Two of them (rs944289 and rs116909374) are located at 14q13 making that locus an important target of research into the genetic predisposition to PTC. We aimed at uncovering other variants at 14q13 associated with PTC independently from the GWAS variants. We performed next generation sequencing of the 14q13 region and analyzed the allele frequencies of single nucleotide polymorphisms (SNPs) in n = 90 PTC cases vs. n = 379 EUR controls from the 1,000 Genome Project. The variants associated with PTC were validated in an Ohio cohort of n = 1,216 PTC cases and n = 1,416 controls. Next, we analyzed the association between SNPs and expression of nearby genes and clinical parameters. We showed that rs368187 was associated with PTC (OR = 1.31, p = 2.20 × 10-6 ). Rs1632250, Rs1863347 and rs1755787 showed association with classical PTC (cPTC) (n = 891; OR = 1.24, 2.22 × 10-3 , OR = 1.31, p = 2.15 × 10-4 and OR = 1.24, p = 2.06 × 10-3 , respectively) while variant rs28397092 showed association with follicular variant (n = 243; OR = 1.51, p = 1.36 × 10-3 ). Rs1863347 was associated with suppression of PTCSC3 in unaffected thyroid tissue (p = 0.026). Rs1632250, rs1863347 and rs1755787 showed association with multifocality (OR = 1.85, p = 0.001, OR = 1.98, p = 0.001 and OR = 1.76, p = 0.003 respectively) and N stage (OR = 1.79, p = 0.014, OR = 1.73, p = 0.023 and OR = 1.81, p = 0.013, respectively) in microPTC (n = 328) while rs368187 was associated with M stage (OR = 0.56, p = 0.034) in cPTC. Our results disclose multiple variants associated with PTC and clinical features in the 14q13 superlocus. We suggest that translational genotype/phenotype studies should take into account not only somatic mutations but also germline variants.
- Published
- 2018
- Full Text
- View/download PDF
33. Genome-wide association study identifies an acute myeloid leukemia susceptibility locus near BICRA
- Author
-
Brian Giacopelli, Markus Scholz, Pamela Brock, Sebastian Schwind, Andrew J. Carroll, Marius Bill, Christopher C. Oakes, John C. Byrd, Dietger Niederwieser, Christopher J. Walker, Richard Stone, Krzysztof Mrózek, Eunice S. Wang, Ann-Kathrin Eisfeld, Jonathan E. Kolitz, Clara D. Bloomfield, Bayard L. Powell, Luke K Genutis, Sandya Liyanarachchi, Albert de la Chapelle, Jessica Kohlschmidt, and Deedra Nicolet
- Subjects
0301 basic medicine ,Genetics ,03 medical and health sciences ,Cancer Research ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Susceptibility locus ,Myeloid leukemia ,Genome-wide association study ,Hematology ,Biology - Published
- 2018
- Full Text
- View/download PDF
34. The role of SMAD3 in the genetic predisposition to papillary thyroid carcinoma
- Author
-
Lianbo Yu, Yanqiang Wang, John E. Phay, Luke K Genutis, Pamela Brock, Rulong Shen, W. G. Li, Sandya Liyanarachchi, Albert de la Chapelle, and Huiling He
- Subjects
0301 basic medicine ,Chromatin Immunoprecipitation ,Locus (genetics) ,Biology ,Polymorphism, Single Nucleotide ,Linkage Disequilibrium ,Article ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Cell Line, Tumor ,medicine ,Humans ,Genetic Predisposition to Disease ,Smad3 Protein ,Thyroid Neoplasms ,Promoter Regions, Genetic ,Thyroid cancer ,Gene ,Genetics (clinical) ,Regulator gene ,Genetics ,Chromosomes, Human, Pair 15 ,Gene knockdown ,Microarray analysis techniques ,Intron ,Chromosome Mapping ,medicine.disease ,030104 developmental biology ,Haplotypes ,Thyroid Cancer, Papillary ,030220 oncology & carcinogenesis ,Chromatin immunoprecipitation ,Protein Binding - Abstract
To identify and characterize the functional variants, regulatory gene networks, and potential binding targets of SMAD3 in the 15q22 thyroid cancer risk locus. We performed linkage disequilibrium (LD) and haplotype analyses to fine map the 15q22 locus. Luciferase reporter assays were applied to evaluate the regulatory effects of the candidate variants. Knockdown by small interfering RNA, microarray analysis, chromatin immunoprecipitation (ChIP) and quantitative real-time polymerase chain reaction assays were performed to reveal the regulatory gene network and identify its binding targets. We report a 25.6-kb haplotype within SMAD3 containing numerous single-nucleotide polymorphisms (SNPs) in high LD. SNPs rs17293632 and rs4562997 were identified as functional variants of SMAD3 by luciferase assays within the LD region. These variants regulate SMAD3 transcription in an allele-specific manner through enhancer elements in introns of SMAD3. Knockdown of SMAD3 in thyroid cancer cell lines revealed its regulatory gene network including two upregulated genes, SPRY4 and SPRY4-IT1. Sequence analysis and ChIP assays validated the actual binding of SMAD3 protein to multiple SMAD binding element sites in the region upstream of SPRY4. Our data provide a functional annotation of the 15q22 thyroid cancer risk locus.
- Published
- 2018
- Full Text
- View/download PDF
35. A Truncating Germline Mutation of
- Author
-
Huiling, He, Wei, Li, Daniel F, Comiskey, Sandya, Liyanarachchi, Taina T, Nieminen, Yanqiang, Wang, Katherine E, DeLap, Pamela, Brock, and Albert, de la Chapelle
- Subjects
Adult ,Aged, 80 and over ,Male ,Telomere-Binding Proteins ,Thyroid Cancer and Nodules ,Middle Aged ,Telomere ,Thyroid Cancer, Papillary ,Adenocarcinoma, Follicular ,Humans ,Female ,Genetic Predisposition to Disease ,Thyroid Neoplasms ,Melanoma ,Germ-Line Mutation - Abstract
Background: Our genome sequencing analysis revealed a frameshift mutation in the shelterin gene TINF2 in a large family with individuals affected with papillary thyroid carcinoma (PTC) and melanoma. Here, we further characterized the mutation and screened for coding variants in the 6 shelterin genes in 24 families. Methods: Sanger sequencing was performed to screen for the TINF2 mutation in the key family. Quantitative reverse transcription-polymerase chain reaction (PCR) was used for TINF2 gene expression analysis. Exogenous expression and co-immunoprecipitation techniques were used for assessing TINF2 binding to TERF1. Relative telomere length (RTL) was quantified in DNAs from lymphocytes by using quantitative real-time PCR. Whole exome sequencing (WES) was performed in seven families with individuals affected with PTC and other cancer types. Screening for DNA variants in shelterin genes was performed by using whole genome sequencing data from 17 families and WES data from 7 further families. Results: The TINF2 mutation (TINF2 p.Trp198fs) showed complete co-segregation with PTC and melanoma in the key family. The mutation is not reported in databases and not identified in 23 other families we screened. The expression of TINF2 was borderline reduced in individuals with the mutation. The truncated TINF2 protein showed abolished binding to TERF1. The RTL in the individuals with the mutation was significantly longer when compared with those without the mutation from the same family as well as compared with 62 healthy controls. Among the 24 families, we identified 3 missense and 1 synonymous variant(s) in 2 shelterin genes (TINF2 and ACD). Conclusions: The rare frameshift mutation in the TINF2 gene and the associated longer telomere length suggest that dysregulated telomeres could be a mechanism predisposing to PTC and melanoma. DNA coding variants in shelterin genes are rare. Further studies are required to evaluate the roles of variants in shelterin genes in thyroid cancer and melanoma.
- Published
- 2020
36. Assessing thyroid cancer risk using polygenic risk scores
- Author
-
Romana T. Netea-Maier, Erich M. Sturgis, Huiling He, Li Xu, Sandya Liyanarachchi, Fadi Nabhan, Egil Ferkingstad, Julius Gudmundsson, Pamela Brock, Gudmar Thorleifsson, Albert de la Chapelle, Matthew D. Ringel, Folkert W. Asselbergs, Theo S. Plantinga, Kari Stefansson, Jose I. Mayordomo, Jon Hrafnkelsson, Hannes Hjartarson, Vinicius Tragante, Jon G. Jonasson, and Lambertus A. Kiemeney
- Subjects
Adult ,Male ,Oncology ,Multifactorial Inheritance ,medicine.medical_specialty ,endocrine system diseases ,DNA Mutational Analysis ,Iceland ,Thyroid Gland ,Penetrance ,Genome-wide association study ,Single-nucleotide polymorphism ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,Polymorphism, Single Nucleotide ,Risk Assessment ,Papillary thyroid cancer ,Cohort Studies ,Decile ,All institutes and research themes of the Radboud University Medical Center ,Predictive Value of Tests ,Risk Factors ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Genetic Predisposition to Disease ,Thyroid Neoplasms ,Genetic risk ,Thyroid cancer ,Genetic association ,Multidisciplinary ,Models, Genetic ,business.industry ,Middle Aged ,Biological Sciences ,medicine.disease ,United Kingdom ,United States ,ROC Curve ,Thyroid Cancer, Papillary ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,Case-Control Studies ,Female ,Polygenic risk score ,business ,Genome-Wide Association Study - Abstract
Genome-wide association studies (GWASs) have identified at least 10 single-nucleotide polymorphisms (SNPs) associated with papillary thyroid cancer (PTC) risk. Most of these SNPs are common variants with small to moderate effect sizes. Here we assessed the combined genetic effects of these variants on PTC risk by using summarized GWAS results to build polygenic risk score (PRS) models in three PTC study groups from Ohio (1,544 patients and 1,593 controls), Iceland (723 patients and 129,556 controls), and the United Kingdom (534 patients and 407,945 controls). A PRS based on the 10 established PTC SNPs showed a stronger predictive power compared with the clinical factors model, with a minimum increase of area under the receiver-operating curve of 5.4 percentage points ( P ≤ 1.0 × 10 −9 ). Adding an extended PRS based on 592,475 common variants did not significantly improve the prediction power compared with the 10-SNP model, suggesting that most of the remaining undiscovered genetic risk in thyroid cancer is due to rare, moderate- to high-penetrance variants rather than to common low-penetrance variants. Based on the 10-SNP PRS, individuals in the top decile group of PRSs have a close to sevenfold greater risk (95% CI, 5.4–8.8) compared with the bottom decile group. In conclusion, PRSs based on a small number of common germline variants emphasize the importance of heritable low-penetrance markers in PTC.
- Published
- 2020
37. GWAS of thyroid stimulating hormone highlights pleiotropic effects and inverse association with thyroid cancer
- Author
-
Leigha Senter, Ben Michael Brumpton, Julius Gudmundsson, Anita Pandit, Peter Arvan, Sandya Liyanarachchi, Matthew D. Ringel, Juha Karjalainen, George Davey Smith, Anne Heidi Skogholt, Layal Chaker, Hannes Hjartarson, Tanmoy Roychowdhury, Albert de la Chapelle, Wei Zhou, Brooke N. Wolford, Kari Stefansson, Ulla T. Schultheiss, Cristen J. Willer, Whitney E. Hornsby, Lars G. Fritsche, Theo S. Plantinga, Sarah E. Graham, Marco Medici, Jonas B. Nielsen, Li Xu, Morgan Oneka, Jose I. Mayordomo, Kristian Hveem, Cintia Eliana Citterio, Serena Sanna, William Overton, Josh Weinstock, Eirin B. Haug, Lambertus A. Kiemeney, Gudmar Thorleifsson, Chad M. Brummett, Michael Boehnke, Mitja I. Kurki, Aarno Palotie, Omer Kabil, Mark J. Daly, Alexander Teumer, Bjørn Olav Åsvold, Peter N. Taylor, Ida Surakka, Erich M. Sturgis, Romana T. Netea-Maier, Silvia Naitza, Maiken Elvestad Gabrielsen, Huiling He, Anne R. Cappola, Jon Hrafnkelsson, Matthew Zawistowski, Humaira Rasheed, Jon G. Jonasson, Epidemiology, Internal Medicine, Institute for Molecular Medicine Finland, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Centre of Excellence in Complex Disease Genetics, Research Programs Unit, Aarno Palotie / Principal Investigator, Genomics of Neurological and Neuropsychiatric Disorders, HUS Helsinki and Uusimaa Hospital District, Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI), Læknadeild (HÍ), Faculty of Medicine (UI), Heilbrigðisvísindasvið (HÍ), School of Health Sciences (UI), Háskóli Íslands, and University of Iceland
- Subjects
0301 basic medicine ,Multifactorial Inheritance ,Goiter ,endocrine system diseases ,medicine.medical_treatment ,Vascular damage Radboud Institute for Health Sciences [Radboudumc 16] ,Thyrotropin ,General Physics and Astronomy ,Genome-wide association study ,02 engineering and technology ,Genome-wide association studies ,THYROID ,Risk Factors ,Thyroid stimulating hormone ,Prevalence ,lcsh:Science ,Thyroid cancer ,Multidisciplinary ,TSH ,Hormónar ,Genetic analysis ,Thyroid ,COMMON VARIANTS ,Genetic Pleiotropy ,purl.org/becyt/ford/3.1 [https] ,Physical Chromosome Mapping ,021001 nanoscience & nanotechnology ,CANCER ,CONGENITAL HYPOTHYROIDISM ,3. Good health ,Phenotype ,medicine.anatomical_structure ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,purl.org/becyt/ford/3 [https] ,Thyroid function ,Erfðarannsóknir ,0210 nano-technology ,hormones, hormone substitutes, and hormone antagonists ,medicine.medical_specialty ,endocrine system ,Science ,Mutation, Missense ,Development ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,THYROGLOBULIN ,All institutes and research themes of the Radboud University Medical Center ,Thyroid-stimulating hormone ,SDG 3 - Good Health and Well-being ,Internal medicine ,Mendelian randomization ,medicine ,Humans ,Genetic Predisposition to Disease ,Thyroid Neoplasms ,GENOME-WIDE ASSOCIATION ,Author Correction ,MALIGNANCY ,RECEPTOR ,HYPERTENSION ,business.industry ,ENDOGENOUS SUBCLINICAL HYPERTHYROIDISM ,SERUM THYROTROPIN CONCENTRATION ,Efnaskipti ,General Chemistry ,Mendelian Randomization Analysis ,medicine.disease ,Metabolism ,GWAS meta-analysis ,030104 developmental biology ,Endocrinology ,Genetic Loci ,lcsh:Q ,Thyroglobulin ,Gene expression ,3111 Biomedicine ,business ,Genome-Wide Association Study - Abstract
Publsiher's version (útgefin grein), Thyroid stimulating hormone (TSH) is critical for normal development and metabolism. To better understand the genetic contribution to TSH levels, we conduct a GWAS meta-analysis at 22.4 million genetic markers in up to 119,715 individuals and identify 74 genome-wide significant loci for TSH, of which 28 are previously unreported. Functional experiments show that the thyroglobulin protein-altering variants P118L and G67S impact thyroglobulin secretion. Phenome-wide association analysis in the UK Biobank demonstrates the pleiotropic effects of TSH-associated variants and a polygenic score for higher TSH levels is associated with a reduced risk of thyroid cancer in the UK Biobank and three other independent studies. Two-sample Mendelian randomization using TSH index variants as instrumental variables suggests a protective effect of higher TSH levels (indicating lower thyroid function) on risk of thyroid cancer and goiter. Our findings highlight the pleiotropic effects of TSH-associated variants on thyroid function and growth of malignant and benign thyroid tumors., We acknowledge the University of Michigan Precision Health Initiative and Medical School Central Biorepository for providing biospecimen storage, management, processing, and distribution services, and the Center for Statistical Genetics in the Department of Biostatistics at the School of Public Health for genotype data curation, imputation, and management in support of this research (for MGI cohort). The biochemical work at the University of Michigan was done with funding from NIH R01 DK-40344. This research has been conducted using the UK Biobank Resource under application number 24460. The HUNT Study is a collaboration between the HUNT Research Centre (Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology), Nord-Trøndelag County Council, Central Norway Regional Health Authority, and the Norwegian Institute of Public Health. The genotyping in HUNT was financed by the National Institutes of Health; University of Michigan; the Research Council of Norway; the Liaison Committee for Education, Research and Innovation in Central Norway; and the Joint Research Committee between St Olav’s Hospital and the Faculty of Medicine and Health Sciences, NTNU. We thank Sean Caron for web development for data sharing. Support for this research was provided by NIH grant DK062370 (M.B.). Ohio State Cohort was supported by NIH P50CA168505 SPORE (M.D.R. and A.d.l.C.). Support for the HUNT and MGI analysis was additionally provided by R35HL135824 (C.J.W.) and R01HL109946 (C.J.W.). W.Z. was supported by the National Human Genome Research Institute of the National Institutes of Health under award number T32HG010464. G.D.S., B.B., H.R. and E.B.H. work in the Medical Research Council Integrative Epidemiology Unit at the University of Bristol MC_UU_00011/1.
- Published
- 2020
- Full Text
- View/download PDF
38. Variants in
- Author
-
Daniel Forrest, Comiskey, Huiling, He, Sandya, Liyanarachchi, Mehek S, Sheikh, Luke K, Genutis, Isabella V, Hendrickson, Lianbo, Yu, Pamela L, Brock, and Albert, de la Chapelle
- Subjects
Genotype ,Mutation, Missense ,Polymorphism, Single Nucleotide ,Linkage Disequilibrium ,Repressor Proteins ,Haplotypes ,Thyroid Cancer, Papillary ,Cell Line, Tumor ,Humans ,Genetic Predisposition to Disease ,Protein Interaction Maps ,Transcriptome ,Alleles ,Genome-Wide Association Study - Abstract
Papillary thyroid carcinoma (PTC) demonstrates high heritability and a low somatic mutation burden relative to other cancers. Therefore, the genetic risk predisposing to PTC is likely due to a combination of low penetrance variants. A recent genome-wide association study revealed the association of PTC with a missense variant, rs6793295, at 3q26 in a gene called Leucine Repeat Rich Containing 34 (We report the mechanisms of PTC risk at 3q26 using a combination of overexpression, mass spectroscopy, knockdown, transcriptome profiling, migration assays and genetic analysis.We observed differential binding of wild-type and missense LRRC34 to RANBP1. Overexpression of missense LRRC34 reduced RanGTP levels and increased apoptosis. We also identified a second linkage disequilibrium (LD) block upstream ofOur study demonstrates two separate mechanisms, one in G protein signalling and the other in transcriptional control, dictating PTC risk at 3q26 using both biochemical and genetic techniques.
- Published
- 2019
39. Identification of Rare Variants Predisposing to Thyroid Cancer
- Author
-
Huiling He, Pamela Brock, Katherine E. Miller, Keiko Akagi, Yanqiang Wang, W. G. Li, Sandya Liyanarachchi, Daniel F. Comiskey, Albert de la Chapelle, Taina T. Nieminen, Daniel C. Koboldt, Katherine E. Delap, and David E. Symer
- Subjects
Male ,Candidate gene ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Biology ,Frameshift mutation ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Endocrinology ,Genetic linkage ,Genetic predisposition ,medicine ,Humans ,Computer Simulation ,Genetic Predisposition to Disease ,Thyroid Neoplasms ,Thyroid cancer ,Germ-Line Mutation ,Aged ,Genetics ,Sanger sequencing ,Whole genome sequencing ,Aged, 80 and over ,Whole Genome Sequencing ,Cancer ,Thyroid Cancer and Nodules ,Middle Aged ,medicine.disease ,Carcinoma, Papillary ,Pedigree ,Thyroid Cancer, Papillary ,030220 oncology & carcinogenesis ,symbols ,Female ,Genome-Wide Association Study - Abstract
Background: Familial non-medullary thyroid cancer (NMTC) accounts for a relatively small proportion of thyroid cancer cases, but it displays strong genetic predisposition. So far, only a few NMTC susceptible genes and low-penetrance variants contributing to NMTC have been described. This study aimed to identify rare germline variants that may predispose individuals to NMTC by sequencing a cohort of 17 NMTC families. Methods: Whole-genome sequencing and genome-wide linkage analysis were performed in 17 NMTC families. MendelScan and BasePlayer were applied to screen germline variants followed by customized filtering. The remaining candidate variants were subsequently validated by Sanger sequencing. A panel of 277 known cancer predisposition genes was also screened in these families. Results: A total of 41 rare coding candidate variants in 40 genes identified by whole-genome sequencing are reported, including 24 missense, five frameshift, five splice change, and seven nonsense variants. Sanger sequencing confirmed all 41 rare variants and proved their co-segregation with NMTC in the extended pedigrees. In silico functional analysis of the candidate genes using Ingenuity Pathway Analysis showed that cancer was the top category of "Diseases and Disorders." Additionally, a targeted search displayed six variants in known cancer predisposition genes, including one frameshift variant and five missense variants. Conclusions: The data identify rare germline variants that may play important roles in NMTC predisposition. It is proposed that in future research including functional characterization, these variants and genes be considered primary candidates for thyroid cancer predisposition.
- Published
- 2019
40. Genome-Wide Expression Screening Discloses Long Noncoding RNAs Involved in Thyroid Carcinogenesis
- Author
-
Pearlly S. Yan, W. G. Li, Sandya Liyanarachchi, Huiling He, Albert de la Chapelle, Pamela Brock, Leigha Senter, Matthew D. Ringel, and Ralf Bundschuh
- Subjects
0301 basic medicine ,endocrine system diseases ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Thyroid Gland ,Bioinformatics ,medicine.disease_cause ,Biochemistry ,Cohort Studies ,0302 clinical medicine ,Endocrinology ,Gene expression ,RNA, Neoplasm ,Thyroid ,Outcome measures ,Prognosis ,Up-Regulation ,medicine.anatomical_structure ,Thyroid Cancer, Papillary ,Lymphatic Metastasis ,030220 oncology & carcinogenesis ,Thyroidectomy ,RNA, Long Noncoding ,Proto-Oncogene Proteins B-raf ,medicine.medical_specialty ,Expert Systems ,Biology ,Thyroid carcinoma ,03 medical and health sciences ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Thyroid Neoplasms ,Genome wide expression ,Neoplasm Staging ,Ohio ,Gene Expression Profiling ,Carcinoma ,Biochemistry (medical) ,Computational Biology ,RNA ,Original Articles ,Carcinoma, Papillary ,Fold change ,030104 developmental biology ,Amino Acid Substitution ,Mutation ,Cancer research ,Lymph Node Excision ,Lymph Nodes ,Carcinogenesis ,Genome-Wide Association Study - Abstract
Long noncoding RNAs (lncRNAs) regulate pathological processes, yet their potential roles in papillary thyroid carcinoma (PTC) are poorly understood.To profile transcriptionally dysregulated lncRNAs in PTC and identify lncRNAs associated with clinicopathological characteristics.We performed RNA sequencing of 12 paired PTC tumors and matched noncancerous tissues and correlated the expression of lncRNAs with clinical parameters. The 2 most significantly dysregulated lncRNAs were studied in an Ohio PTC cohort (n = 109) and in PTC data (n = 497) from The Cancer Genome Atlas.A combination of laboratory-based studies and computational analysis using clinical data and samples and a publically available database.Correlation between expression values and clinical parameters.We identified 218 lncRNAs showing differential expression in PTC (fold change ≥ 2.0, P.01). Significant correlation was observed between the expression of 2 lncRNAs (XLOC_051122 and XLOC_006074) and 1) lymph node metastasis (N stage) and 2) BRAF(V600E) mutation. Among patients with wild-type BRAF, the expression of these 2 lncRNAs showed significantly higher levels in the patients with lymph node metastasis. In silico analysis of these lncRNAs pinpointed cell movement and cellular growth and proliferation as targeted functions.Comprehensive expression screening identified 2 novel lncRNAs associated with risk factors of adverse prognosis in PTC patients. These lncRNAs may be novel players in PTC carcinogenesis.
- Published
- 2016
- Full Text
- View/download PDF
41. Dissection of the Major Hematopoietic Quantitative Trait Locus in Chromosome 6q23.3 Identifies miR-3662 as a Player in Hematopoiesis and Acute Myeloid Leukemia
- Author
-
Kevin W. Hoag, Ramiro Garzon, Sophia E. Maharry, Christopher J. Walker, Sujay Mehta, Mitra Patel, Denis C. Guttridge, Ann-Kathrin Eisfeld, Xiaomeng Huang, Sandya Liyanarachchi, Albert de la Chapelle, James S. Blachly, Maryam Bainazar, Malori A. Lankenau, Clara D. Bloomfield, and Parvathi Ranganathan
- Subjects
0301 basic medicine ,Myeloid ,Gene Dosage ,Genome-wide association study ,Mice ,0302 clinical medicine ,GATA1 Transcription Factor ,Genetics ,Gene Expression Regulation, Leukemic ,NF-kappa B ,Myeloid leukemia ,I-kappa B Kinase ,Leukemia, Myeloid, Acute ,Leukemia ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Heterografts ,Chromosomes, Human, Pair 6 ,Female ,RNA Interference ,Protein Binding ,Signal Transduction ,Cell Survival ,Quantitative Trait Loci ,Locus (genetics) ,Quantitative trait locus ,Biology ,Response Elements ,Models, Biological ,Polymorphism, Single Nucleotide ,Article ,Colony-Forming Units Assay ,03 medical and health sciences ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Allele ,Enhancer ,Alleles ,Genetic Association Studies ,Cell Proliferation ,Binding Sites ,Hematopoietic Stem Cells ,medicine.disease ,Hematopoiesis ,Disease Models, Animal ,MicroRNAs ,030104 developmental biology ,CCAAT-Enhancer-Binding Proteins ,Genome-Wide Association Study - Abstract
Chromosomal aberrations and multiple genome-wide association studies (GWAS) have established a major hematopoietic quantitative trait locus in chromosome 6q23.3. The locus comprises an active enhancer region, in which some of the associated SNPs alter transcription factor binding. We now identify miR-3662 as a new functional driver contributing to the associated phenotypes. The GWAS SNPs are strongly associated with higher miR-3662 expression. Genome editing of rs66650371, a three-base-pair deletion, suggests a functional link between the SNP genotype and the abundance of miR-3662. Increasing miR-3662′s abundance increases colony formation in hematopoietic progenitor cells, particularly the erythroid lineage. In contrast, miR-3662 is not expressed in acute myeloid leukemia cells, and its overexpression has potent antileukemic effects in vitro and in vivo. Mechanistically, miR-3662 directly targets NF-κB–mediated transcription. Thus, miR-3662 is a new player of the hematopoietic 6q23.3 locus. Significance: The characterization of miR-3662 has identified a new actor in the prominent hematopoietic quantitative trait locus in chromosome 6q23.3. The mechanistic insights into miR-3662′s function may reveal novel or only partially known pathways for normal and malignant hematopoietic cell proliferation. Cancer Discov; 6(9); 1036–51. ©2016 AACR. This article is highlighted in the In This Issue feature, p. 932
- Published
- 2016
- Full Text
- View/download PDF
42. NF-κB inhibition rescues cardiac function by remodeling calcium genes in a Duchenne muscular dystrophy model
- Author
-
Huating Wang, Jin Mo Gu, Jennifer M. Peterson, Sandya Liyanarachchi, Christopher E. Gaw, Vikram Shettigar, Mark T. Ziolo, Benjamin D. Canan, Jennifer M. Petrosino, Steve R. Roof, Sudarshana M. Sharma, Denis C. Guttridge, Nadine Bakkar, David J. Wang, Leina Lu, Paul M.L. Janssen, Jonathan P. Davis, Sean C. Little, Priya Londhe, Jonathan Shintaku, and Nivedita M. Ratnam
- Subjects
Male ,musculoskeletal diseases ,0301 basic medicine ,CCCTC-Binding Factor ,Science ,Duchenne muscular dystrophy ,Cardiomyopathy ,General Physics and Astronomy ,Histone Deacetylase 1 ,Article ,Sodium-Calcium Exchanger ,General Biochemistry, Genetics and Molecular Biology ,Muscle hypertrophy ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Medicine ,Myocyte ,Myocytes, Cardiac ,Muscular dystrophy ,lcsh:Science ,Cells, Cultured ,Multidisciplinary ,business.industry ,NF-kappa B ,Skeletal muscle ,General Chemistry ,Chromatin Assembly and Disassembly ,medicine.disease ,HDAC1 ,Cell biology ,Chromatin ,Muscular Dystrophy, Duchenne ,Repressor Proteins ,Sin3 Histone Deacetylase and Corepressor Complex ,030104 developmental biology ,medicine.anatomical_structure ,Mice, Inbred mdx ,Calcium ,lcsh:Q ,business ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Duchenne muscular dystrophy (DMD) is a neuromuscular disorder causing progressive muscle degeneration. Although cardiomyopathy is a leading mortality cause in DMD patients, the mechanisms underlying heart failure are not well understood. Previously, we showed that NF-κB exacerbates DMD skeletal muscle pathology by promoting inflammation and impairing new muscle growth. Here, we show that NF-κB is activated in murine dystrophic (mdx) hearts, and that cardiomyocyte ablation of NF-κB rescues cardiac function. This physiological improvement is associated with a signature of upregulated calcium genes, coinciding with global enrichment of permissive H3K27 acetylation chromatin marks and depletion of the transcriptional repressors CCCTC-binding factor, SIN3 transcription regulator family member A, and histone deacetylase 1. In this respect, in DMD hearts, NF-κB acts differently from its established role as a transcriptional activator, instead promoting global changes in the chromatin landscape to regulate calcium genes and cardiac function., The molecular mechanisms leading to heart failure in patients with Duchenne muscular dystrophy are unclear. Here the authors show that NF-κB is activated in the heart of dystrophin-deficient mice and that its ablation rescues cardiac function through chromatin remodeling and activation of gene expression.
- Published
- 2018
- Full Text
- View/download PDF
43. Genome-wide association study identifies an acute myeloid leukemia susceptibility locus near BICRA
- Author
-
Christopher J, Walker, Christopher C, Oakes, Luke K, Genutis, Brian, Giacopelli, Sandya, Liyanarachchi, Deedra, Nicolet, Ann-Kathrin, Eisfeld, Markus, Scholz, Pamela, Brock, Jessica, Kohlschmidt, Krzysztof, Mrózek, Marius, Bill, Andrew J, Carroll, Jonathan E, Kolitz, Bayard L, Powell, Eunice S, Wang, Dietger W, Niederwieser, Richard M, Stone, John C, Byrd, Sebastian, Schwind, Albert, de la Chapelle, and Clara D, Bloomfield
- Subjects
Leukemia, Myeloid, Acute ,genome-wide association study ,Genotype ,Genetic Loci ,Chromosomal Proteins, Non-Histone ,Tumor Suppressor Proteins ,Humans ,Genetic Predisposition to Disease ,acute myeloid leukemia ,Polymorphism, Single Nucleotide ,Alleles ,Article ,Cell Line - Published
- 2018
44. PTCSC3 Is Involved in Papillary Thyroid Carcinoma Development by ModulatingS100A4Gene Expression
- Author
-
Jarosław Jendrzejewski, Andrew Eiterman, W. G. Li, Sandya Liyanarachchi, Hanna S. Radomska, Huiling He, Albert de la Chapelle, Krzysztof Sworczak, Rebecca Nagy, Andrew Thomas, and Jerneja Tomsic
- Subjects
Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,RNA, Untranslated ,Genotype ,Carcinogenesis ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Thyroid Gland ,Down-Regulation ,Context (language use) ,Biology ,medicine.disease_cause ,Biochemistry ,Thyroid carcinoma ,Endocrinology ,Cell Line, Tumor ,Internal medicine ,Gene expression ,medicine ,Humans ,Genetic Predisposition to Disease ,Neoplasm Invasiveness ,S100 Calcium-Binding Protein A4 ,Thyroid Neoplasms ,Cell Proliferation ,Regulation of gene expression ,Matrigel ,JCEM Online: Advances in Genetics ,S100 Proteins ,Biochemistry (medical) ,Molecular biology ,Carcinoma, Papillary ,Gene Expression Regulation, Neoplastic ,Reverse transcription polymerase chain reaction ,Real-time polymerase chain reaction ,Matrix Metalloproteinase 9 - Abstract
We previously showed that a long noncoding RNA gene, PTCSC3, located close to the variant rs944289 that predisposes to papillary thyroid carcinoma (PTC) might target the S100A4 gene.The aim was to investigate the impact of PTCSC3 on S100A4 expression and its role in cancer development.S100A4 abundance was analyzed by quantitative PCR (qPCR) in unaffected and tumor tissue from n = 73 PTC patients. The expression of PTCSC3 and S100A4 was studied in BCPAP and TPC-1 cell lines with forced expression of PTCSC3 by qPCR. Expression of S100A4 target genes (VEGF and MMP-9) was studied in the BCPAP cell line with forced expression of PTCSC3 by qPCR, reverse transcriptase PCR, and Western blot. The impact of PTCSC3 on BCPAP motility and invasiveness was analyzed by the Transwell and Matrigel assays, respectively.This was a laboratory-based study using cells from clinical samples and thyroid cancer cell lines.We aimed to find evidence for a link between the expression of PTCSC3 and thyroid carcinogenesis.Expression data from PTC cell lines pinpointed S100A4 as the most significantly downregulated gene in the presence of PTCSC3. S100A4 was upregulated in tumor tissue (P = 9.33 × 10(-7)) while PTCSC3 was strongly downregulated (P = 2.2 × 10(-16)). S100A4 transcription was moderately correlated with PTCSC3 expression in unaffected thyroid tissue (r = 0.429, P = .0001), and strongly in unaffected tissue of patients with the risk allele of rs944289 (r = 0.685, P = 7.88 × 10(-5)). S100A4, VEGF, and MMP-9 were suppressed in the presence of PTCSC3 (P = .0051, P = .0090, and P =.0037, respectively). PTC cells expressing PTCSC3 showed reduction in motility and invasiveness (P = 4.52 × 10(-5) and P = 1.0 × 10(-4), respectively).PTCSC3 downregulates S100A4, leading to a reduction in cell motility and invasiveness. We propose that PTCSC3 impacts PTC predisposition and carcinogenesis through the S100A4 pathway.
- Published
- 2015
- Full Text
- View/download PDF
45. Multiple functional variants in long-range enhancer elements contribute to the risk of SNP rs965513 in thyroid cancer
- Author
-
Yao Wang, Huiling He, Yanqiang Wang, Mukund Srinivas, Rebecca Nagy, Dayong Wu, Qianben Wang, Victor X. Jin, John E. Phay, Rulong Shen, David E. Symer, W. G. Li, Sandya Liyanarachchi, Albert de la Chapelle, and Keiko Akagi
- Subjects
Chromatin Immunoprecipitation ,Genotype ,endocrine system diseases ,Penetrance ,Single-nucleotide polymorphism ,Genome-wide association study ,Biology ,Polymorphism, Single Nucleotide ,Papillary thyroid cancer ,Histones ,Thyroid carcinoma ,Chromosome conformation capture ,Cell Line, Tumor ,Odds Ratio ,medicine ,Humans ,Genetic Predisposition to Disease ,Thyroid Neoplasms ,Enhancer ,Thyroid cancer ,Alleles ,Genetics ,Multidisciplinary ,Carcinoma ,Forkhead Transcription Factors ,Biological Sciences ,medicine.disease ,Carcinoma, Papillary ,Chromatin ,Enhancer Elements, Genetic ,Haplotypes ,Thyroid Cancer, Papillary ,Genome-Wide Association Study ,FOXE1 - Abstract
The [A] allele of SNP rs965513 in 9q22 has been consistently shown to be highly associated with increased papillary thyroid cancer (PTC) risk with an odds ratio of ∼1.8 as determined by genome-wide association studies, yet the molecular mechanisms remain poorly understood. Previously, we noted that the expression of two genes in the region, forkhead box E1 (FOXE1) and PTC susceptibility candidate 2 (PTCSC2), is regulated by rs965513 in unaffected thyroid tissue, but the underlying mechanisms were not elucidated. Here, we fine-mapped the 9q22 region in PTC and controls and detected an ∼33-kb linkage disequilibrium block (containing the lead SNP rs965513) that significantly associates with PTC risk. Chromatin characteristics and regulatory element signatures in this block disclosed at least three regulatory elements functioning as enhancers. These enhancers harbor at least four SNPs (rs7864322, rs12352658, rs7847449, and rs10759944) that serve as functional variants. The variant genotypes are associated with differential enhancer activities and/or transcription factor binding activities. Using the chromosome conformation capture methodology, long-range looping interactions of these elements with the promoter region shared by FOXE1 and PTCSC2 in a human papillary thyroid carcinoma cell line (KTC-1) and unaffected thyroid tissue were found. Our results suggest that multiple variants coinherited with the lead SNP and located in long-range enhancers are involved in the transcriptional regulation of FOXE1 and PTCSC2 expression. These results explain the mechanism by which the risk allele of rs965513 predisposes to thyroid cancer.
- Published
- 2015
- Full Text
- View/download PDF
46. Genetic Predisposition to Papillary Thyroid Carcinoma: Involvement of FOXE1, TSHR, and a Novel lincRNA Gene, PTCSC2
- Author
-
Huiling He, Rebecca Nagy, Ramana V. Davuluri, Mukund Srinivas, Jarosław Jendrzejewski, W. G. Li, Sandya Liyanarachchi, and Albert de la Chapelle
- Subjects
Genotype ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Locus (genetics) ,Biology ,Polymorphism, Single Nucleotide ,Biochemistry ,Thyroid carcinoma ,Endocrinology ,Rapid amplification of cDNA ends ,Gene expression ,Genetic predisposition ,medicine ,Humans ,Genetic Predisposition to Disease ,Thyroid Neoplasms ,Gene ,Alleles ,JCEM Online: Advances in Genetics ,Biochemistry (medical) ,Thyroid ,Forkhead Transcription Factors ,Receptors, Thyrotropin ,Molecular biology ,Carcinoma, Papillary ,medicine.anatomical_structure ,RNA, Long Noncoding ,Genome-Wide Association Study ,FOXE1 - Abstract
Context: By genome-wide association studies, the risk allele [A] of SNP rs965513 predisposes strongly to papillary thyroid carcinoma (PTC). It is located in a gene-poor region of 9q22, some 60 kb from the FOXE1 gene. The underlying mechanisms remain to be discovered. Objective: Our objective was to identify novel transcripts in the 9q22 locus and correlate gene expression levels with the genotypes of rs965513. Design: We performed 3′ and 5′ rapid amplification of cDNA ends and RT-PCR to detect novel transcripts. One novel transcript was forcibly expressed in a cell line followed by gene expression array analysis. We genotyped rs965513 from PTC patients and measured gene expression levels by real-time RT-PCR in unaffected thyroid tissue and matched tumor. Setting: This was a laboratory-based study using cells from clinical tissue samples and a cancer cell line. Main Outcome Measures: We detected previously uncharacterized transcripts and evaluated the gene expression levels and the correlation with the risk allele of rs965513, age, gender, chronic lymphocyte thyroiditis (CLT), and TSH levels. Results: We found a novel long intergenic noncoding RNA gene and named it papillary thyroid cancer susceptibility candidate 2 (PTCSC2). Transcripts of PTCSC2 are down-regulated in PTC tumors. The risk allele [A] of rs965513 was significantly associated with low expression of unspliced PTCSC2, FOXE1, and TSHR in unaffected thyroid tissue. We also observed a significant association of age and CLT with PTCSC2 unspliced transcript levels. The correlation between the rs965513 genotype and the PTCSC2 unspliced transcript levels remained significant after adjusting for age, gender, and CLT. Forced expression of PTCSC2 in the BCPAP cell line affected the expression of a subset of noncoding and coding transcripts with enrichment of genes functionally involved in cell cycle and cancer. Conclusions: Our data suggest a role for PTCSC2, FOXE1, and TSHR in the predisposition to PTC.
- Published
- 2015
- Full Text
- View/download PDF
47. Penetrance of a rare familial mutation predisposing to papillary thyroid cancer
- Author
-
Pamela Brock, Jennifer A. Sipos, Albert de la Chapelle, Donika Saporito, Heather Hampel, Soledad Fernandez, Rebecca Nagy, and Sandya Liyanarachchi
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,030209 endocrinology & metabolism ,Penetrance ,Article ,Papillary thyroid cancer ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Internal medicine ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Thyroid Neoplasms ,Anaplastic thyroid cancer ,Family history ,Thyroid cancer ,Genetics (clinical) ,Germ-Line Mutation ,Aged ,business.industry ,Thyroid disease ,Thyroid ,Middle Aged ,medicine.disease ,Pedigree ,medicine.anatomical_structure ,Thyroid Cancer, Papillary ,030220 oncology & carcinogenesis ,Mutation (genetic algorithm) ,Female ,business - Abstract
Familial non-medullary thyroid cancer (FNMTC) is clinically defined as two or more first-degree relatives with NMTC and appears to follow an autosomal dominant inheritance pattern. Approximately 5–7% of NMTC is hereditary and affects multiple generations with a young age of onset. The primary aim of this study was to determine the age-specific penetrance of NMTC in individuals from a large family with FNMTC with a previously identified private mutation at 4q32, with a secondary aim to determine the penetrance for benign thyroid disease in this family. We present a large family with NMTC in which we had previously described a culpable mutation. Participants provided their personal medical history and family history. The germline 4q32 A > C mutation was detected in 34 of 68 tested individuals. Age-specific penetrance of thyroid cancer and benign thyroid disease was determined using the inverted Kaplan–Meier method of segregation analysis. Individuals who tested positive for the 4q32 mutation have a 68.9% (95% CI 46.5–88.7) risk of developing thyroid cancer by age 70 and a 65.3% (95% CI 46.0–83.8) risk of developing benign thyroid disease by age 70. The 4q32 A > C mutation significantly increases the risk to develop thyroid cancer but not benign thyroid disease in members of this family. The female:male sex ratio of 1.33 that we observed in affected mutation carriers differs greatly from the ratio of approximately 3:1 observed in PTC, supporting a central role of the mutation. Early thyroid surveillance with annual ultrasound is recommended to individuals testing positive for this private familial mutation.
- Published
- 2017
48. NRAS isoforms differentially affect downstream pathways, cell growth, and cell transformation
- Author
-
Sebastian Schwind, Xiaomeng Huang, Sandya Liyanarachchi, Wenrui Duan, Albert de la Chapelle, William E. Carson, Ann-Kathrin Eisfeld, Kevin W. Hoag, Clara D. Bloomfield, Ravi Patel, Christopher J. Walker, Guido Marcucci, Joseph Markowitz, and Gregory A. Otterson
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Gene isoform ,viruses ,Blotting, Western ,Molecular Sequence Data ,Biology ,Statistics, Nonparametric ,GTP Phosphohydrolases ,Mice ,Carcinoma, Non-Small-Cell Lung ,Chlorocebus aethiops ,Animals ,Humans ,Immunoprecipitation ,Protein Isoforms ,Small GTPase ,Cloning, Molecular ,DNA Primers ,Regulation of gene expression ,Microscopy, Confocal ,Multidisciplinary ,Base Sequence ,Cell growth ,Membrane Proteins ,Sequence Analysis, DNA ,Biological Sciences ,Molecular biology ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,Ras Signaling Pathway ,Cytoplasm ,COS Cells ,NIH 3T3 Cells ,Signal transduction ,Signal Transduction - Abstract
Neuroblastoma rat sarcoma (RAS) viral oncogene homolog (NRAS), a small GTPase, is one of the most thoroughly studied oncogenes that controls cell growth, differentiation, and survival by facilitating signal transduction. Here, we identify four novel naturally occurring NRAS isoforms (isoforms 2-5) in addition to the canonical isoform (isoform 1). Expression analyses performed on a panel of several different human malignancies and matching normal tissue revealed distinct isoform expression patterns. Two of the novel isoforms were found in the nucleus and cytoplasm, whereas the others were exclusively cytoplasmic. The isoforms varied in their binding affinities to known downstream targets and differentially regulated the RAS signaling pathway. Strikingly, forced expression of isoform 5, which encodes only a 20-aa peptide, led to increased cell proliferation and to transformation by activation of known NRAS targets. These discoveries open new avenues in the study of NRAS.
- Published
- 2014
- Full Text
- View/download PDF
49. Abstract 4156: Multiple rare germline variants in a thyroid cancer family
- Author
-
Taina T. Nieminen, Daniel F. Comiskey, Yanqiang Wang, Sandya Liyanarachchi, Pamela Brock, Paivi Peltomaki, Huiling He, and Albert de la Chapelle
- Subjects
Cancer Research ,Oncology - Abstract
Thyroid cancer is the 12th most common cancer in the USA. Four main types exist: the majority (85-90%) is papillary thyroid cancer (PTC). Familial form of PTC represents 5 to 15 % of all thyroid cancers. We have access to 155 PTC families including clinical data and biological samples. Our aim was to search for highly penetrant gene variants in 17 PTC families. Data from one of the families in which PTC is segregating in three generations are described here. Whole genome sequencing was performed in three family members affected by PTC. Sanger sequencing at germline DNA and RNA levels followed by TOPO cloning was used to study the effects of mutations. We found 4 different either novel or ultra-rare mutations in the ITGAD, AARS and PDPR genes on chromosome 16 (Table). All mutations segregated perfectly with PTC in family members (n=4) and were absent in unaffected members (n=3). We Sanger sequenced cDNA from two of the PTC patients and cloned the products into the pCR 2.1-® TOPO vector. Based on the cDNA sequencing and cloning results, all of the mutations will create a premature stop codon leading to a truncated protein. We will verify the effects of the mutations by protein analysis as well. In addition, structural variation analysis will be done to exclude large genomic rearrangements. The germline events leading to PTC are neither well known nor extensively researched. When occurring in families PTC regularly shows autosomal dominant inheritance with incomplete penetrance. Only a few of probable mutations have been reported. Nevertheless, our finding of the truncating mutations in 3 genes on chromosome 16 that are cosegregating with PTC in a 3-generation family is surprising. Even more so: is the localization of the three genes in one chromosome coincidental? Preliminary findings in some of the other 16 PTC families suggest that the occurrence of more than one truncating mutation is not a unique event. We look forward to answer the question whether combination of two or more mutant genes can be responsible for one disease segregating in families. Citation Format: Taina T. Nieminen, Daniel F. Comiskey, Yanqiang Wang, Sandya Liyanarachchi, Pamela Brock, Paivi Peltomaki, Huiling He, Albert de la Chapelle. Multiple rare germline variants in a thyroid cancer family [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4156.
- Published
- 2019
- Full Text
- View/download PDF
50. Recurrent and founder mutations in thePMS2gene
- Author
-
Leigha Senter, John L. Hopper, Mark Clendenning, Wade S. Samowitz, Joanne P. Young, Jerneja Tomsic, Cecily P. Vaughn, A de la Chapelle, Mark A. Jenkins, and Sandya Liyanarachchi
- Subjects
Genetics ,congenital, hereditary, and neonatal diseases and abnormalities ,Mutation ,Pseudogene ,Haplotype ,Biology ,medicine.disease ,medicine.disease_cause ,MLH1 ,Lynch syndrome ,Germline mutation ,Genotype ,medicine ,Genetics (clinical) ,Founder effect - Abstract
Germline mutations in PMS2 are associated with Lynch syndrome (LS), the most common known cause of hereditary colorectal cancer. Mutation detection in PMS2 has been difficult due to the presence of several pseudogenes, but a custom-designed long-range PCR strategy now allows adequate mutation detection. Many mutations are unique. However, some mutations are observed repeatedly across individuals not known to be related due to the mutation being either recurrent, arising multiple times de novo at hot spots for mutations, or of founder origin, having occurred once in an ancestor. Previously, we observed 36 distinct mutations in a sample of 61 independently ascertained Caucasian probands of mixed European background with PMS2 mutations. Eleven of these mutations were detected in more than one individual not known to be related and of these, six were detected more than twice. These six mutations accounted for 31 (51%) ostensibly unrelated probands. Here, we performed genotyping and haplotype analysis in four mutations observed in multiple probands and found two (c.137G>T and exon 10 deletion) to be founder mutations and one (c.903G>T) a probable founder. One (c.1A>G) could not be evaluated for founder mutation status. We discuss possible explanations for the frequent occurrence of founder mutations in PMS2.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.